1
|
Wang Z, Zhang G, Hu S, Fu M, Zhang P, Zhang K, Hao L, Chen S. Research progress on the protective effect of hormones and hormone drugs in myocardial ischemia-reperfusion injury. Biomed Pharmacother 2024; 176:116764. [PMID: 38805965 DOI: 10.1016/j.biopha.2024.116764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 05/05/2024] [Accepted: 05/17/2024] [Indexed: 05/30/2024] Open
Abstract
Ischemic heart disease (IHD) is a condition where the heart muscle does not receive enough blood flow, leading to cardiac dysfunction. Restoring blood flow to the coronary artery is an effective clinical therapy for myocardial ischemia. This strategy helps lower the size of the myocardial infarction and improves the prognosis of patients. Nevertheless, if the disrupted blood flow to the heart muscle is restored within a specific timeframe, it leads to more severe harm to the previously deprived heart tissue. This condition is referred to as myocardial ischemia/reperfusion injury (MIRI). Until now, there is a dearth of efficacious strategies to prevent and manage MIRI. Hormones are specialized substances that are produced directly into the circulation by endocrine organs or tissues in humans and animals, and they have particular effects on the body. Hormonal medications utilize human or animal hormones as their active components, encompassing sex hormones, adrenaline medications, thyroid hormone medications, and others. While several studies have examined the preventive properties of different endocrine hormones, such as estrogen and hormone analogs, on myocardial injury caused by ischemia-reperfusion, there are other hormone analogs whose mechanisms of action remain unexplained and whose safety cannot be assured. The current study is on hormones and hormone medications, elucidating the mechanism of hormone pharmaceuticals and emphasizing the cardioprotective effects of different endocrine hormones. It aims to provide guidance for the therapeutic use of drugs and offer direction for the examination of MIRI in clinical therapy.
Collapse
Affiliation(s)
- Zhongyi Wang
- Department of Pharmaceutical Toxicology, School of Pharmacy, China Medical University, Shenyang, 110122, China
| | - Gaojiang Zhang
- Department of Pharmaceutical Toxicology, School of Pharmacy, China Medical University, Shenyang, 110122, China
| | - Shan Hu
- Department of Pharmaceutical Toxicology, School of Pharmacy, China Medical University, Shenyang, 110122, China
| | - Meilin Fu
- Department of Pharmaceutical Toxicology, School of Pharmacy, China Medical University, Shenyang, 110122, China
| | - Pingyuan Zhang
- Department of Pharmaceutical Toxicology, School of Pharmacy, China Medical University, Shenyang, 110122, China
| | - Kuo Zhang
- Department of Pharmaceutical Toxicology, School of Pharmacy, China Medical University, Shenyang, 110122, China
| | - Liying Hao
- Department of Pharmaceutical Toxicology, School of Pharmacy, China Medical University, Shenyang, 110122, China.
| | - Sichong Chen
- Department of Pharmaceutical Toxicology, School of Pharmacy, China Medical University, Shenyang, 110122, China.
| |
Collapse
|
2
|
Bai Y, Zhao F, Wu T, Chen F, Pang X. Actin polymerization and depolymerization in developing vertebrates. Front Physiol 2023; 14:1213668. [PMID: 37745245 PMCID: PMC10515290 DOI: 10.3389/fphys.2023.1213668] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 08/22/2023] [Indexed: 09/26/2023] Open
Abstract
Development is a complex process that occurs throughout the life cycle. F-actin, a major component of the cytoskeleton, is essential for the morphogenesis of tissues and organs during development. F-actin is formed by the polymerization of G-actin, and the dynamic balance of polymerization and depolymerization ensures proper cellular function. Disruption of this balance results in various abnormalities and defects or even embryonic lethality. Here, we reviewed recent findings on the structure of G-actin and F-actin and the polymerization of G-actin to F-actin. We also focused on the functions of actin isoforms and the underlying mechanisms of actin polymerization/depolymerization in cellular and organic morphogenesis during development. This information will extend our understanding of the role of actin polymerization in the physiologic or pathologic processes during development and may open new avenues for developing therapeutics for embryonic developmental abnormalities or tissue regeneration.
Collapse
Affiliation(s)
- Yang Bai
- Stomatological Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Feng Zhao
- Stomatological Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Tingting Wu
- Stomatological Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Fangchun Chen
- Stomatological Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Xiaoxiao Pang
- Stomatological Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| |
Collapse
|
3
|
Healing the Broken Hearts: A Glimpse on Next Generation Therapeutics. HEARTS 2022. [DOI: 10.3390/hearts3040013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Cardiovascular diseases are the leading cause of death worldwide, accounting for 32% of deaths globally and thus representing almost 18 million people according to WHO. Myocardial infarction, the most prevalent adult cardiovascular pathology, affects over half a million people in the USA according to the last records of the AHA. However, not only adult cardiovascular diseases are the most frequent diseases in adulthood, but congenital heart diseases also affect 0.8–1.2% of all births, accounting for mild developmental defects such as atrial septal defects to life-threatening pathologies such as tetralogy of Fallot or permanent common trunk that, if not surgically corrected in early postnatal days, they are incompatible with life. Therefore, both congenital and adult cardiovascular diseases represent an enormous social and economic burden that invariably demands continuous efforts to understand the causes of such cardiovascular defects and develop innovative strategies to correct and/or palliate them. In the next paragraphs, we aim to briefly account for our current understanding of the cellular bases of both congenital and adult cardiovascular diseases, providing a perspective of the plausible lines of action that might eventually result in increasing our understanding of cardiovascular diseases. This analysis will come out with the building blocks for designing novel and innovative therapeutic approaches to healing the broken hearts.
Collapse
|
4
|
Maselli D, Matos RS, Johnson RD, Chiappini C, Camelliti P, Campagnolo P. Epicardial slices: an innovative 3D organotypic model to study epicardial cell physiology and activation. NPJ Regen Med 2022; 7:7. [PMID: 35039552 PMCID: PMC8764051 DOI: 10.1038/s41536-021-00202-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Accepted: 11/30/2021] [Indexed: 11/26/2022] Open
Abstract
The epicardium constitutes an untapped reservoir for cardiac regeneration. Upon heart injury, the adult epicardium re-activates, leading to epithelial-to-mesenchymal transition (EMT), migration, and differentiation. While interesting mechanistic and therapeutic findings arose from lower vertebrates and rodent models, the introduction of an experimental system representative of large mammals would undoubtedly facilitate translational advancements. Here, we apply innovative protocols to obtain living 3D organotypic epicardial slices from porcine hearts, encompassing the epicardial/myocardial interface. In culture, our slices preserve the in vivo architecture and functionality, presenting a continuous epicardium overlaying a healthy and connected myocardium. Upon thymosin β4 treatment of the slices, the epicardial cells become activated, upregulating epicardial and EMT genes, resulting in epicardial cell mobilization and differentiation into epicardial-derived mesenchymal cells. Our 3D organotypic model enables to investigate the reparative potential of the adult epicardium, offering an advanced tool to explore ex vivo the complex 3D interactions occurring within the native heart environment.
Collapse
Affiliation(s)
- D Maselli
- Faculty of Health & Medical Sciences, School of Biosciences & Medicine, Section of Cardiovascular Sciences, University of Surrey, Guildford, GU2 7XH, UK
| | - R S Matos
- Faculty of Health & Medical Sciences, School of Biosciences & Medicine, Section of Cardiovascular Sciences, University of Surrey, Guildford, GU2 7XH, UK
| | - R D Johnson
- Faculty of Health & Medical Sciences, School of Biosciences & Medicine, Section of Cardiovascular Sciences, University of Surrey, Guildford, GU2 7XH, UK
| | - C Chiappini
- Centre for Craniofacial and Regenerative Biology, King's College London, SE1 9RT, London, United Kingdom
| | - P Camelliti
- Faculty of Health & Medical Sciences, School of Biosciences & Medicine, Section of Cardiovascular Sciences, University of Surrey, Guildford, GU2 7XH, UK
| | - P Campagnolo
- Faculty of Health & Medical Sciences, School of Biosciences & Medicine, Section of Cardiovascular Sciences, University of Surrey, Guildford, GU2 7XH, UK.
| |
Collapse
|
5
|
Zhao K, Yang CX, Li P, Sun W, Kong XQ. Epigenetic role of N6-methyladenosine (m6A) RNA methylation in the cardiovascular system. J Zhejiang Univ Sci B 2021; 21:509-523. [PMID: 32633106 DOI: 10.1631/jzus.b1900680] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
As the most prevalent and abundant transcriptional modification in the eukaryotic genome, the continuous and dynamic regulation of N6-methyladenosine (m6A) has been shown to play a vital role in physiological and pathological processes of cardiovascular diseases (CVDs), such as ischemic heart failure (HF), myocardial hypertrophy, myocardial infarction (MI), and cardiomyogenesis. Regulation is achieved by modulating the expression of m6A enzymes and their downstream cardiac genes. In addition, this process has a major impact on different aspects of internal biological metabolism and several other external environmental effects associated with the development of CVDs. However, the exact molecular mechanism of m6A epigenetic regulation has not been fully elucidated. In this review, we outline recent advances and discuss potential therapeutic strategies for managing m6A in relation to several common CVD-related metabolic disorders and external environmental factors. Note that an appropriate understanding of the biological function of m6A in the cardiovascular system will pave the way towards exploring the mechanisms responsible for the development of other CVDs and their associated symptoms. Finally, it can provide new insights for the development of novel therapeutic agents for use in clinical practice.
Collapse
Affiliation(s)
- Kun Zhao
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Chuan-Xi Yang
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Peng Li
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Wei Sun
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Xiang-Qing Kong
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| |
Collapse
|
6
|
Maar K, Hetenyi R, Maar S, Faskerti G, Hanna D, Lippai B, Takatsy A, Bock-Marquette I. 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; 10:1343. [PMID: 34071596 PMCID: PMC8228050 DOI: 10.3390/cells10061343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [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
Affiliation(s)
- Klaudia Maar
- Department of Biochemistry and Medical Chemistry, Medical School, University of Pecs, Szigeti Street 12., H-7624 Pecs, Hungary; (K.M.); (R.H.); (S.M.); (G.F.); (D.H.); (A.T.)
- Szentagothai Research Centre, Research Group of Regenerative Science, Sport and Medicine, University of Pecs, Ifjusag Street 20. C301, H-7624 Pecs, Hungary;
| | - Roland Hetenyi
- Department of Biochemistry and Medical Chemistry, Medical School, University of Pecs, Szigeti Street 12., H-7624 Pecs, Hungary; (K.M.); (R.H.); (S.M.); (G.F.); (D.H.); (A.T.)
- Szentagothai Research Centre, Research Group of Regenerative Science, Sport and Medicine, University of Pecs, Ifjusag Street 20. C301, H-7624 Pecs, Hungary;
| | - Szabolcs Maar
- Department of Biochemistry and Medical Chemistry, Medical School, University of Pecs, Szigeti Street 12., H-7624 Pecs, Hungary; (K.M.); (R.H.); (S.M.); (G.F.); (D.H.); (A.T.)
- Szentagothai Research Centre, Research Group of Regenerative Science, Sport and Medicine, University of Pecs, Ifjusag Street 20. C301, H-7624 Pecs, Hungary;
| | - Gabor Faskerti
- Department of Biochemistry and Medical Chemistry, Medical School, University of Pecs, Szigeti Street 12., H-7624 Pecs, Hungary; (K.M.); (R.H.); (S.M.); (G.F.); (D.H.); (A.T.)
- Szentagothai Research Centre, Research Group of Regenerative Science, Sport and Medicine, University of Pecs, Ifjusag Street 20. C301, H-7624 Pecs, Hungary;
| | - Daniel Hanna
- Department of Biochemistry and Medical Chemistry, Medical School, University of Pecs, Szigeti Street 12., H-7624 Pecs, Hungary; (K.M.); (R.H.); (S.M.); (G.F.); (D.H.); (A.T.)
- Szentagothai Research Centre, Research Group of Regenerative Science, Sport and Medicine, University of Pecs, Ifjusag Street 20. C301, H-7624 Pecs, Hungary;
| | - Balint Lippai
- Szentagothai Research Centre, Research Group of Regenerative Science, Sport and Medicine, University of Pecs, Ifjusag Street 20. C301, H-7624 Pecs, Hungary;
| | - Aniko Takatsy
- Department of Biochemistry and Medical Chemistry, Medical School, University of Pecs, Szigeti Street 12., H-7624 Pecs, Hungary; (K.M.); (R.H.); (S.M.); (G.F.); (D.H.); (A.T.)
- Szentagothai Research Centre, Research Group of Regenerative Science, Sport and Medicine, University of Pecs, Ifjusag Street 20. C301, H-7624 Pecs, Hungary;
| | - Ildiko Bock-Marquette
- Department of Biochemistry and Medical Chemistry, Medical School, University of Pecs, Szigeti Street 12., H-7624 Pecs, Hungary; (K.M.); (R.H.); (S.M.); (G.F.); (D.H.); (A.T.)
- Szentagothai Research Centre, Research Group of Regenerative Science, Sport and Medicine, University of Pecs, Ifjusag Street 20. C301, H-7624 Pecs, Hungary;
| |
Collapse
|
7
|
MRI-based molecular imaging of epicardium-derived stromal cells (EpiSC) by peptide-mediated active targeting. Sci Rep 2020; 10:21669. [PMID: 33303866 PMCID: PMC7728754 DOI: 10.1038/s41598-020-78600-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 11/20/2020] [Indexed: 12/20/2022] Open
Abstract
After myocardial infarction (MI), epicardial cells reactivate their embryonic program, proliferate and migrate into the damaged tissue to differentiate into fibroblasts, endothelial cells and, if adequately stimulated, to cardiomyocytes. Targeting epicardium-derived stromal cells (EpiSC) by specific ligands might enable the direct imaging of EpiSCs after MI to better understand their biology, but also may permit the cell-specific delivery of small molecules to improve the post-MI healing process. Therefore, the aim of this study was to identify specific peptides by phage display screening to enable EpiSC specific cargo delivery by active targeting. To this end, we utilized a sequential panning of a phage library on cultured rat EpiSCs and then subtracted phage that nonspecifically bound blood immune cells. EpiSC specific phage were analyzed by deep sequencing and bioinformatics analysis to identify a total of 78 300 ± 31 900 different, EpiSC-specific, peptide insertion sequences. Flow cytometry of the five most highly abundant peptides (EP1, -2, -3, -7 or EP9) showed strong binding to EpiSCs but not to blood immune cells. The best binding properties were found for EP9 which was further studied by surface plasmon resonance (SPR). SPR revealed rapid and stable association of EpiSCs with EP9. As a negative control, THP-1 monocytes did not associate with EP9. Coupling of EP9 to perfluorocarbon nanoemulsions (PFCs) resulted in the efficient delivery of 19F cargo to EpiSCs and enabled their visualization by 19F MRI. Moreover, active targeting of EpiSCs by EP9-labelled PFCs was able to outcompete the strong phagocytic uptake of PFCs by circulating monocytes. In summary, we have identified a 7-mer peptide, (EP9) that binds to EpiSCs with high affinity and specificity. This peptide can be used to deliver small molecule cargos such as contrast agents to permit future in vivo tracking of EpiSCs by molecular imaging and to transfer small pharmaceutical molecules to modulate the biological activity of EpiSCs.
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: 2.4] [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
|
Abstract
The heart is lined by a single layer of mesothelial cells called the epicardium that provides important cellular contributions for embryonic heart formation. The epicardium harbors a population of progenitor cells that undergo epithelial-to-mesenchymal transition displaying characteristic conversion of planar epithelial cells into multipolar and invasive mesenchymal cells before differentiating into nonmyocyte cardiac lineages, such as vascular smooth muscle cells, pericytes, and fibroblasts. The epicardium is also a source of paracrine cues that are essential for fetal cardiac growth, coronary vessel patterning, and regenerative heart repair. Although the epicardium becomes dormant after birth, cardiac injury reactivates developmental gene programs that stimulate epithelial-to-mesenchymal transition; however, it is not clear how the epicardium contributes to disease progression or repair in the adult. In this review, we will summarize the molecular mechanisms that control epicardium-derived progenitor cell migration, and the functional contributions of the epicardium to heart formation and cardiomyopathy. Future perspectives will be presented to highlight emerging therapeutic strategies aimed at harnessing the regenerative potential of the fetal epicardium for cardiac repair.
Collapse
Affiliation(s)
- Pearl Quijada
- From the Aab Cardiovascular Research Institute (P.Q., E.M.S.), University of Rochester, School of Medicine and Dentistry, Rochester, NY.,Department of Medicine (P.Q., E.M.S.), University of Rochester, School of Medicine and Dentistry, Rochester, NY
| | | | - Eric M Small
- From the Aab Cardiovascular Research Institute (P.Q., E.M.S.), University of Rochester, School of Medicine and Dentistry, Rochester, NY.,Department of Medicine (P.Q., E.M.S.), University of Rochester, School of Medicine and Dentistry, Rochester, NY
| |
Collapse
|
10
|
Scalise M, Marino F, Cianflone E, Mancuso T, Marotta P, Aquila I, Torella M, Nadal-Ginard B, Torella D. Heterogeneity of Adult Cardiac Stem Cells. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1169:141-178. [PMID: 31487023 DOI: 10.1007/978-3-030-24108-7_8] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Cardiac biology and heart regeneration have been intensively investigated and debated in the last 15 years. Nowadays, the well-established and old dogma that the adult heart lacks of any myocyte-regenerative capacity has been firmly overturned by the evidence of cardiomyocyte renewal throughout the mammalian life as part of normal organ cell homeostasis, which is increased in response to injury. Concurrently, reproducible evidences from independent laboratories have convincingly shown that the adult heart possesses a pool of multipotent cardiac stem/progenitor cells (CSCs or CPCs) capable of sustaining cardiomyocyte and vascular tissue refreshment after injury. CSC transplantation in animal models displays an effective regenerative potential and may be helpful to treat chronic heart failure (CHF), obviating at the poor/modest results using non-cardiac cells in clinical trials. Nevertheless, the degree/significance of cardiomyocyte turnover in the adult heart, which is insufficient to regenerate extensive damage from ischemic and non-ischemic origin, remains strongly disputed. Concurrently, different methodologies used to detect CSCs in situ have created the paradox of the adult heart harboring more than seven different cardiac progenitor populations. The latter was likely secondary to the intrinsic heterogeneity of any regenerative cell agent in an adult tissue but also to the confusion created by the heterogeneity of the cell population identified by a single cell marker used to detect the CSCs in situ. On the other hand, some recent studies using genetic fate mapping strategies claimed that CSCs are an irrelevant endogenous source of new cardiomyocytes in the adult. On the basis of these contradictory findings, here we critically reviewed the available data on adult CSC biology and their role in myocardial cell homeostasis and repair.
Collapse
Affiliation(s)
- Mariangela Scalise
- Molecular and Cellular Cardiology Laboratory, Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy
| | - Fabiola Marino
- Molecular and Cellular Cardiology Laboratory, Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy
| | - Eleonora Cianflone
- Molecular and Cellular Cardiology Laboratory, Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy
| | - Teresa Mancuso
- Molecular and Cellular Cardiology Laboratory, Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy
| | - Pina Marotta
- Molecular and Cellular Cardiology Laboratory, Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy
| | - Iolanda Aquila
- Molecular and Cellular Cardiology Laboratory, Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy
| | - Michele Torella
- Department of Cardiothoracic Surgery, University of Campania "L.Vanvitelli", Naples, Italy
| | - Bernardo Nadal-Ginard
- Molecular and Cellular Cardiology Laboratory, Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy
| | - Daniele Torella
- Molecular and Cellular Cardiology Laboratory, Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy.
| |
Collapse
|
11
|
Huang Z, Song Y, Pang Z, Zhang B, Yang H, Shi H, Chen J, Gong H, Qian J, Ge J. Targeted delivery of thymosin beta 4 to the injured myocardium using CREKA-conjugated nanoparticles. Int J Nanomedicine 2017; 12:3023-3036. [PMID: 28442910 PMCID: PMC5396927 DOI: 10.2147/ijn.s131949] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Purpose Thymosin beta 4 (Tβ4) has multiple beneficial facets for myocardial injury, but its efficiency is limited by the low local concentration within the infarct. Here, we established a Tβ4 delivery system for cardiac repair based on the interaction between the abundant fibrin in the infarct zone and the fibrin-targeting moiety clot-binding peptide cysteine–arginine–glutamic acid–lysine–alanine (CREKA). Methods and results CREKA and Tβ4 were conjugated to nanoparticles (CNP–Tβ4). In vitro binding test revealed that CNP–Tβ4 had a significant binding ability to the surface of fibrin clots when compared to the control clots (NP–Tβ4). Based on the validation of fibrin expression in the early stage of ischemia injury, CNP–Tβ4 was intravenously administered to mice with acute myocardial ischemia–reperfusion injury. CNP–Tβ4 revealed a stronger fibrin-targeting ability than the NP–Tβ4 group and accumulated mainly in the infarcted area and colocalized with fibrin. Subsequently, treatment with CNP–Tβ4 resulted in a better therapeutic effect. Conclusion CRKEA modification favored Tβ4 accumulation and retention in the infarcted region, leading to augmented functional benefits. Fibrin-targeting delivery system represents a generalizable platform technology for regenerative medicine.
Collapse
Affiliation(s)
- Zheyong Huang
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University
| | - Yanan Song
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University
| | - Zhiqing Pang
- School of Pharmacy, Fudan University, Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai
| | - Bo Zhang
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, Hubei
| | - Hongbo Yang
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University
| | - Hongtao Shi
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University
| | - Jing Chen
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University
| | - Hui Gong
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University.,Institute of Biomedical Science, Fudan University, Shanghai, People's Republic of China
| | - Juying Qian
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University
| | - Junbo Ge
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University.,Institute of Biomedical Science, Fudan University, Shanghai, People's Republic of China
| |
Collapse
|
12
|
Abstract
Treatment with thymosin beta 4 (Tβ4) reduces infarct volume and preserves cardiac function in preclinical models of cardiac ischemic injury. These effects stem in part from decreased infarct size, but additional benefits are likely due to specific antifibrotic and proangiogenic activities. Injected or transgenic Tβ4 increase blood vessel growth in large and small animal models, consistent with Tβ4 converting hibernating myocardium to an actively contractile state following ischemia. Tβ4 and its degradation products have antifibrotic effects in in vitro assays and in animal models of fibrosis not related to cardiac injury. This large number of pleiotropic effects results from Tβ4's many interactions with cellular signaling pathways, particularly indirect regulation of cellular motility and movement via the SRF-MRTF-G-actin transcriptional pathway. Variation in effects and effect sizes in animal models may potentially be due to variable distribution of Tβ4. Preclinical studies of PK/PD relationships and a reliable pharmacodynamic biomarker would facilitate clinical development of Tβ4.
Collapse
Affiliation(s)
- G T Pipes
- Cardiovascular Drug Discovery, Discovery Biology Research & Development, Bristol-Myers Squibb, Pennington, NJ, United States.
| | - J Yang
- Cardiovascular Drug Discovery, Discovery Biology Research & Development, Bristol-Myers Squibb, Pennington, NJ, United States
| |
Collapse
|
13
|
Zhang J, Zhang ZG, Li Y, Lu M, Zhang Y, Elias SB, Chopp M. Thymosin beta4 promotes oligodendrogenesis in the demyelinating central nervous system. Neurobiol Dis 2016; 88:85-95. [DOI: 10.1016/j.nbd.2016.01.010] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Revised: 12/19/2015] [Accepted: 01/09/2016] [Indexed: 02/01/2023] Open
|
14
|
Abstract
The spatiotemporal expression pattern of Wt1 has been extensively studied in a number of animal models to establish its function and the developmental fate of the cells expressing this gene. In this chapter, we review the available animal models for Wt1-expressing cell lineage analysis, including direct Wt1 expression reporters and systems for permanent Wt1 lineage tracing. We describe the presently used constitutive or inducible genetic lineage tracing approaches based on the Cre/loxP system utilizing Cre recombinase expression under control of a Wt1 promoter.To make these systems accessible, we provide laboratory protocols that include dissection and processing of the tissues for immunofluorescence and histopathological analysis of the lineage-labeled Wt1-derived cells within the embryo/tissue context.
Collapse
Affiliation(s)
- Bettina Wilm
- Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, UK.
| | - Ramon Muñoz-Chapuli
- Department of Animal Biology, Faculty of Science, University of Malaga, Malaga, Spain.
| |
Collapse
|
15
|
Hinkel R, Ball HL, DiMaio JM, Shrivastava S, Thatcher JE, Singh AN, Sun X, Faskerti G, Olson EN, Kupatt C, Bock-Marquette I. C-terminal variable AGES domain of Thymosin β4: the molecule's primary contribution in support of post-ischemic cardiac function and repair. J Mol Cell Cardiol 2015; 87:113-25. [PMID: 26255251 DOI: 10.1016/j.yjmcc.2015.07.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Revised: 06/12/2015] [Accepted: 07/08/2015] [Indexed: 12/19/2022]
Abstract
Repairing defective cardiac cells is important towards improving heart function. Due to the frequency and severity of ischemic heart disease, management of patients featuring this type of cardiac failure receives significant interest. Previously we discovered that Thymosin β4 (TB4), a 43 amino-acid secreted actin sequestering peptide, is beneficial for myocardial cell survival and coronary re-growth after infarction in adult mammals. Considering the regenerative potential of full-length TB4 in the heart, and that minimal structural variations alter TB4's influence on actin assembly and cell movement, we investigated how various TB4 domains affect cardiac cell behavior and post-ischemic mammalian heart function. We synthesized 17 domain combinations of full-length TB4 and analyzed their impact on embryonic cardiac cells in vitro, and after cardiac infarction in vivo. We discovered the domains of TB4 affect cardiac cell behavior distinctly. We revealed TB4 specific C-terminal tetrapeptide, AGES, increases embryonic cardiac cell migration and myocyte beating in culture, and improves adult mammalian heart function following ischemia. Investigating the molecular background and mechanism we discovered systemic injection of AGES enhances early myocyte survival by activating Akt-mediated signaling mechanisms, increases coronary vessel growth and inhibits inflammation in mice and pigs. Biodistribution analyses revealed cardiomyocytes uptake AGES efficiently in vitro and in vivo projecting a potential independent clinical utilization for the tetrapeptide. Our comprehensive domain investigations also suggest, preservation and/or restoration of cardiomyocyte communication is a target of TB4 and AGES, and critical to improve post-ischemic heart function in pigs. In summary, we identified the C-terminal four amino-acid variable end of TB4 as the essential and responsible domain for the molecule's full benefits in the hypoxic heart. Additionally, we introduced AGES as a novel, systemically applicable drug candidate to aid cardiac infarction in adult mammals.
Collapse
Affiliation(s)
- Rabea Hinkel
- Internal Medicine I, University Clinic Grosshadern, Munich 81377, Germany
| | - Haydn L Ball
- Protein Chemistry Technology Center University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - J Michael DiMaio
- Department of Cardiovascular and Thoracic Surgery University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Santwana Shrivastava
- Department of Cardiovascular and Thoracic Surgery University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Jeffrey E Thatcher
- Department of Cardiovascular and Thoracic Surgery University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Ajay N Singh
- Advanced Imaging Research Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Xiankai Sun
- Advanced Imaging Research Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Gabor Faskerti
- University of Pecs, Faculty of Medicine, Szentagothai Research Centre, Pecs 7624, Hungary
| | - Eric N Olson
- Department of Molecular Biology University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Christian Kupatt
- Internal Medicine I, University Clinic Grosshadern, Munich 81377, Germany
| | - Ildiko Bock-Marquette
- Department of Cardiovascular and Thoracic Surgery University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; University of Pecs, Faculty of Medicine, Szentagothai Research Centre, Pecs 7624, Hungary.
| |
Collapse
|
16
|
Choudry FA, Yeo C, Mozid A, Martin JF, Mathur A. Increases in plasma Tβ4 after intracardiac cell therapy in chronic ischemic heart failure is associated with symptomatic improvement. Regen Med 2015; 10:403-10. [DOI: 10.2217/rme.15.9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Aim: Tβ4 is an integral factor in repair of myocardium in animal models. To investigate whether Tβ4 is important in human cardiac disease and has a role in mediating the beneficial cardiac effects of bone-marrow-derived stem cell (BMSC) therapy, we measured serial plasma Tβ4 levels in patients enrolled on the REGENERATE-IHD cell therapy trial. Patients & Methods: Plasma Tβ4 concentrations were measured in 13 patients who received BMSCs and 14 controls. Results: There was a significant increase in plasma Tβ4 in the BMSC group 24 h after intracardiac injection. Increases in Tβ4 levels were associated with improvement in New York Heart Association symptom class. Conclusion: This exploratory study highlights the need for further study of Tβ4 in human cardiovascular disease.
Collapse
Affiliation(s)
- Fizzah A Choudry
- Department of Cardiology, London Chest Hospital, Barts Health Trust, London, E2 9JX, UK
| | - Chia Yeo
- Department of Cardiology, London Chest Hospital, Barts Health Trust, London, E2 9JX, UK
| | - Abdul Mozid
- Department of Cardiology, London Chest Hospital, Barts Health Trust, London, E2 9JX, UK
| | - John F Martin
- British Heart Foundation Laboratories, Department of Medicine, University College London, London, WC1E 6JJ, UK
| | - Anthony Mathur
- Department of Cardiology, London Chest Hospital, Barts Health Trust, London, E2 9JX, UK
- Centre for Clinical Pharmacology, Cardiovascular Biomedical Research Unit, The William Harvey Research Institute, Queen Mary University of London, London, EC1M 6BQ, UK
| |
Collapse
|
17
|
Moon JH, Lee JW, Kang JK. Angiogenic effects of recombinant thymosin β4 in a mouse hindlimb ischemia model. J Biomed Res 2014. [DOI: 10.12729/jbr.2014.15.4.156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
18
|
Abstract
During development, cardiogenesis is orchestrated by a family of heart progenitors that build distinct regions of the heart. Each region contains diverse cell types that assemble to form the complex structures of the individual cardiac compartments. Cardiomyocytes are the main cell type found in the heart and ensure contraction of the chambers and efficient blood flow throughout the body. Injury to the cardiac muscle often leads to heart failure due to the loss of a large number of cardiomyocytes and its limited intrinsic capacity to regenerate the damaged tissue, making it one of the leading causes of morbidity and mortality worldwide. In this Primer we discuss how insights into the molecular and cellular framework underlying cardiac development can be used to guide the in vitro specification of cardiomyocytes, whether by directed differentiation of pluripotent stem cells or via direct lineage conversion. Additional strategies to generate cardiomyocytes in situ, such as reactivation of endogenous cardiac progenitors and induction of cardiomyocyte proliferation, will also be discussed.
Collapse
Affiliation(s)
- Daniela Später
- Department of Stem Cell and Regenerative Biology, Harvard University and Harvard Medical School, 7 Divinity Avenue, Cambridge, MA 02138, USA Department of Bioscience, CVMD iMED, AstraZeneca, Pepparedsleden 1, Mölndal 43150, Sweden
| | - Emil M Hansson
- Department of Stem Cell and Regenerative Biology, Harvard University and Harvard Medical School, 7 Divinity Avenue, Cambridge, MA 02138, USA Department of Cell and Molecular Biology and Medicine, Karolinska Institutet, 35 Berzelius Vag, Stockholm 171 77, Sweden
| | - Lior Zangi
- Department of Stem Cell and Regenerative Biology, Harvard University and Harvard Medical School, 7 Divinity Avenue, Cambridge, MA 02138, USA Department of Cardiology, Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, USA Cardiovascular Research Center, Mount Sinai School of Medicine, One Gustave L. Levy Place, New York, NY 10029, USA
| | - Kenneth R Chien
- Department of Stem Cell and Regenerative Biology, Harvard University and Harvard Medical School, 7 Divinity Avenue, Cambridge, MA 02138, USA Department of Cell and Molecular Biology and Medicine, Karolinska Institutet, 35 Berzelius Vag, Stockholm 171 77, Sweden
| |
Collapse
|
19
|
Adeno-Associated Viral Vector 2.9 Thymosin ß4 Application Attenuates Rejection After Heart Transplantation. Transplantation 2014; 98:835-43. [DOI: 10.1097/tp.0000000000000327] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
20
|
Abstract
Human heart failure (HF) is one of the leading causes of morbidity and mortality worldwide. Currently, heart transplantation and implantation of mechanical devices represent the only available treatments for advanced HF. Two alternative strategies have emerged to treat patients with HF. One approach relies on transplantation of exogenous stem cells (SCs) of non-cardiac or cardiac origin to induce cardiac regeneration and improve ventricular function. Another complementary strategy relies on stimulation of the endogenous regenerative capacity of uninjured cardiac progenitor cells to rebuild cardiac muscle and restore ventricular function. Various SC types and delivery strategies have been examined in the experimental and clinical settings; however, neither the ideal cell type nor the cell delivery method for cardiac cell therapy has yet emerged. Although the use of bone marrow (BM)-derived cells, most frequently exploited in clinical trials, appears to be safe, the results are controversial. Two recent randomized trials have failed to document any beneficial effects of intracardiac delivery of autologous BM mononuclear cells on cardiac function of patients with HF. The remarkable discovery that various populations of cardiac progenitor cells (CPCs) are present in the adult human heart and that it possesses limited regeneration capacity has opened a new era in cardiac repair. Importantly, unlike BM-derived SCs, autologous CPCs from myocardial biopsies cultured and subsequently delivered by coronary injection to patients have given positive results. Although these data are promising, a better understanding of how to control proliferation and differentiation of CPCs, to enhance their recruitment and survival, is required before CPCs become clinically applicable therapeutics.
Collapse
Affiliation(s)
- Alexander T Akhmedov
- The Molecular Cardiology and Neuromuscular Institute, 75 Raritan Ave., Highland Park, NJ, 08904, USA
| | | |
Collapse
|
21
|
Bollini S, Vieira JMN, Howard S, Dubè KN, Balmer GM, Smart N, Riley PR. Re-activated adult epicardial progenitor cells are a heterogeneous population molecularly distinct from their embryonic counterparts. Stem Cells Dev 2014; 23:1719-30. [PMID: 24702282 DOI: 10.1089/scd.2014.0019] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Cardiovascular disease remains the major cause of mortality, and cardiac cell therapy has recently emerged as a paradigm for heart repair. The epicardium is a layer of mesothelial cells covering the heart that during development contributes to different cardiovascular lineages, including cardiomyocytes, but which becomes quiescent after birth. We previously revealed that the peptide thymosin beta 4 (Tβ4) can reactivate adult epicardium-derived cells (EPDCs) after myocardial infarction (MI), to proliferate, and differentiate into cardiovascular derivatives. The aim of this study was to provide a lineage characterization of the adult EPDCs relative to the embryonic epicardial lineage and to determine prospective cell fate biases within the activated adult population during cardiovascular repair. Wt1(GFPCre/+) mice were primed with Tβ4 and MI induced by ligation of the left anterior descending coronary artery. Adult WT1(+) GFP(+) EPDCs were fluorescence-activated cell sorted (FACS) at 2, 4, and 7 days after MI. Embryonic WT1(+) GFP(+) EPDCs were isolated from embryonic hearts (E12.5) by FACS, and sorted cells were characterized by real-time quantitative reverse transcriptase-polymerase chain reaction (RT-qPCR) and immunostaining. Adult WT1(+) GFP(+) EPDCs were highly heterogeneous, expressing cardiac progenitor and mesenchymal stem markers. Based on the expression of stem cell antigen-1 (Sca-1), CD44, and CD90, we identified different subpopulations of EPDCs of varying cardiovascular potential, according to marker gene profiles, with a molecular phenotype distinct from the source embryonic epicardial cells at E12.5. Thus, adult WT1(+) GFP(+) cells are a heterogeneous population that when activated can restore an embryonic gene programme, but do not revert entirely to adopt an embryonic phenotype. Potential biases in cardiovascular cell fate suggest that discrete subpopulations of EPDCs might be clinically relevant for regenerative therapy.
Collapse
Affiliation(s)
- Sveva Bollini
- 1 Department of Physiology, Anatomy and Genetics, University of Oxford , Oxford, United Kingdom
| | | | | | | | | | | | | |
Collapse
|
22
|
Andersen DC, Ganesalingam S, Jensen CH, Sheikh SP. Do neonatal mouse hearts regenerate following heart apex resection? Stem Cell Reports 2014; 2:406-13. [PMID: 24749066 PMCID: PMC3986579 DOI: 10.1016/j.stemcr.2014.02.008] [Citation(s) in RCA: 93] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2013] [Revised: 02/24/2014] [Accepted: 02/25/2014] [Indexed: 11/30/2022] Open
Abstract
The mammalian heart has generally been considered nonregenerative, but recent progress suggests that neonatal mouse hearts have a genuine capacity to regenerate following apex resection (AR). However, in this study, we performed AR or sham surgery on 400 neonatal mice from inbred and outbred strains and found no evidence of complete regeneration. Ideally, new functional cardiomyocytes, endothelial cells, and vascular smooth muscle cells should be formed in the necrotic area of the damaged heart. Here, damaged hearts were 9.8% shorter and weighed 14% less than sham controls. In addition, the resection border contained a massive fibrotic scar mainly composed of nonmyocytes and collagen disposition. Furthermore, there was a substantial reduction in the number of proliferating cardiomyocytes in AR hearts. Our results thus question the usefulness of the AR model for identifying molecular mechanisms underlying regeneration of the adult heart after damage.
Collapse
Affiliation(s)
- Ditte Caroline Andersen
- Laboratory of Molecular and Cellular Cardiology, Department of Clinical Biochemistry and Pharmacology, Odense University Hospital, Winsloewparken 21 , 5000 Odense C, Denmark ; Clinical Institute, University of Southern Denmark, 5000 Odense C, Denmark
| | - Suganya Ganesalingam
- Laboratory of Molecular and Cellular Cardiology, Department of Clinical Biochemistry and Pharmacology, Odense University Hospital, Winsloewparken 21 , 5000 Odense C, Denmark ; Institute of Molecular Medicine, University of Southern Denmark, 5000 Odense C, Denmark
| | - Charlotte Harken Jensen
- Laboratory of Molecular and Cellular Cardiology, Department of Clinical Biochemistry and Pharmacology, Odense University Hospital, Winsloewparken 21 , 5000 Odense C, Denmark
| | - Søren Paludan Sheikh
- Laboratory of Molecular and Cellular Cardiology, Department of Clinical Biochemistry and Pharmacology, Odense University Hospital, Winsloewparken 21 , 5000 Odense C, Denmark ; Institute of Molecular Medicine, University of Southern Denmark, 5000 Odense C, Denmark
| |
Collapse
|
23
|
Michalak K, Czesny S, Epifanio J, Snyder RJ, Schultz ET, Velotta JP, McCormick SD, Brown BL, Santopietro G, Michalak P. Beta-thymosin gene polymorphism associated with freshwater invasiveness of alewife (Alosa pseudoharengus). ACTA ACUST UNITED AC 2014; 321:233-40. [DOI: 10.1002/jez.1854] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2013] [Revised: 11/17/2013] [Accepted: 01/06/2014] [Indexed: 02/01/2023]
Affiliation(s)
| | - Sergiusz Czesny
- Lake Michigan Biological Station; Illinois Natural History Survey; Prairie Research Institute; University of Illinois; Champaign Illinois
| | - John Epifanio
- Illinois Natural History Survey; Prairie Research Institute; Champaign Illinois
| | | | - Eric T. Schultz
- Department of Ecology and Evolutionary Biology; University of Connecticut; Storrs Connecticut
| | - Jonathan P. Velotta
- Department of Ecology and Evolutionary Biology; University of Connecticut; Storrs Connecticut
| | | | - Bonnie L. Brown
- Department of Biology; Virginia Commonwealth University; Richmond Virginia
| | | | - Pawel Michalak
- Virginia Bioinformatics Institute; Virginia Tech; Blacksburg Virginia
| |
Collapse
|
24
|
Chankiewitz V, Morosan-Puopolo G, Yusuf F, Rudloff S, Pröls F, Kleff V, Hofmann DK, Brand-Saberi B. A thymosin beta15-like peptide promotes intersegmental myotome extension in the chicken embryo. Histochem Cell Biol 2013; 141:275-87. [DOI: 10.1007/s00418-013-1156-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/06/2013] [Indexed: 12/21/2022]
|
25
|
Yan B, Singla RD, Abdelli LS, Singal PK, Singla DK. Regulation of PTEN/Akt pathway enhances cardiomyogenesis and attenuates adverse left ventricular remodeling following thymosin β4 Overexpressing embryonic stem cell transplantation in the infarcted heart. PLoS One 2013; 8:e75580. [PMID: 24086577 PMCID: PMC3782449 DOI: 10.1371/journal.pone.0075580] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2013] [Accepted: 08/13/2013] [Indexed: 11/19/2022] Open
Abstract
Thymosin β4 (Tβ4), a small G-actin sequestering peptide, mediates cell proliferation, migration, and angiogenesis. Whether embryonic stem (ES) cells, overexpressing Tβ4, readily differentiate into cardiac myocytes in vitro and in vivo and enhance cardioprotection following transplantation post myocardial infarction (MI) remains unknown. Accordingly, we established stable mouse ES cell lines, RFP-ESCs and Tβ4-ESCs, expressing RFP and an RFP-Tβ4 fusion protein, respectively. In vitro, the number of spontaneously beating embryoid bodies (EBs) was significantly increased in Tβ4-ESCs at day 9, 12 and 15, compared with RFP-ESCs. Enhanced expression of cardiac transcriptional factors GATA-4, Mef2c and Txb6 in Tβ4-EBs, as confirmed with real time-PCR analysis, was accompanied by the increased number of EB areas stained positive for sarcomeric α-actin in Tβ4-EBs, compared with the RFP control, suggesting a significant increase in functional cardiac myocytes. Furthermore, we transplanted Tβ4-ESCs into the infarcted mouse heart and performed morphological and functional analysis 2 weeks after MI. There was a significant increase in newly formed cardiac myocytes associated with the Notch pathway, a decrease in apoptotic nuclei mediated by an increase in Akt and a decrease in levels of PTEN. Cardiac fibrosis was significantly reduced, and left ventricular function was significantly augmented in the Tβ4-ESC transplanted group, compared with controls. It is concluded that genetically modified Tβ4-ESCs, potentiates their ability to turn into cardiac myocytes in vitro as well as in vivo. Moreover, we also demonstrate that there was a significant decrease in both cardiac apoptosis and fibrosis, thus improving cardiac function in the infarcted heart.
Collapse
Affiliation(s)
- Binbin Yan
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, Florida, United States of America
| | - Reetu D. Singla
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, Florida, United States of America
| | - Latifa S. Abdelli
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, Florida, United States of America
| | - Pawan K. Singal
- Institute of Cardiovascular Sciences, St. Boniface General Hospital Research Centre, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Dinender K. Singla
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, Florida, United States of America
- * E-mail:
| |
Collapse
|
26
|
Tang YL, Wang YJ, Chen LJ, Pan YH, Zhang L, Weintraub NL. Cardiac-derived stem cell-based therapy for heart failure: progress and clinical applications. Exp Biol Med (Maywood) 2013; 238:294-300. [PMID: 23598975 DOI: 10.1177/1535370213477982] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Stem cell-based therapy is emerging as a promising strategy to treat end-stage heart failure, a leading cause of morbidity and mortality. Stem cells can be isolated from a variety of sources and exhibit unique characteristics that impact their potential therapeutic utility. The adult heart contains small populations of committed, multipotent cardiac stem cells (CSC), which are adapted to the cardiac microenvironment and participate in postnatal physiological and pathological cardiac renewal or repair. These cells can be isolated, expanded in culture, and administered therapeutically to improve cardiac function in the setting of heart failure. CSC can be differentiated into three distinct cardiovascular lineages and exhibit enhanced paracrine factor production and engraftment as compared with other types of mesenchymal stem cells, which in turn may translate into improved therapeutic efficacy. The cell surface marker expression and phenotype of these CSC, however, depends on the method of isolation, selection and propagation, which likely explains the variable experimental results obtained to date. Moreover, invasive procedures are required to obtain CSC from humans. Early trials using autologous CSC in patients with ischemic cardiomyopathy have demonstrated feasibility and safety, along with variable degrees of therapeutic efficacy in terms of enhancing myocardial viability and cardiac function. Further studies are needed to optimize methods of CSC isolation, manipulation and delivery. If fully realized, the potential of CSC therapy could fundamentally change the approach to the treatment of end-stage heart failure.
Collapse
Affiliation(s)
- Yaoliang L Tang
- Division of Cardiovascular Disease, Department of Internal Medicine, College of Medicine, University of Cincinnati, Ohio 45267, USA.
| | | | | | | | | | | |
Collapse
|
27
|
Tenorio-Laranga J, Peltonen I, Keskitalo S, Duran-Torres G, Natarajan R, Männistö PT, Nurmi A, Vartiainen N, Airas L, Elovaara I, García-Horsman JA. Alteration of prolyl oligopeptidase and activated α-2-macroglobulin in multiple sclerosis subtypes and in the clinically isolated syndrome. Biochem Pharmacol 2013; 85:1783-94. [PMID: 23643808 DOI: 10.1016/j.bcp.2013.04.018] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2013] [Revised: 04/19/2013] [Accepted: 04/23/2013] [Indexed: 01/24/2023]
Abstract
Prolyl oligopeptidase (PREP) has been considered as a drug target for the treatment of neurodegenerative diseases. In plasma, PREP has been found altered in several disorders of the central nervous system including multiple sclerosis (MS). Oxidative stress and the levels of an endogenous plasma PREP inhibitor have been proposed to decrease PREP activity in MS. In this work, we measured the circulating levels of PREP in patients suffering of relapsing remitting (RR), secondary progressive (SP), primary progressive (PP) MS, and in subjects with clinically isolated syndrome (CIS). We found a significantly lower PREP activity in plasma of RRMS as well as in PPMS patients and a trend to reduced activity in subjects diagnosed with CIS, compared to controls. No signs of oxidative inactivation of PREP, and no correlation with the endogenous PREP inhibitor, identified as activated α-2-macroglobulin (α2M*), were observed in any of the patients studied. However, a significant decrease of α2M* was recorded in MS. In cell cultures, we found that PREP specifically stimulates immune active cells possibly by modifying the levels of fibrinogen β, thymosin β4, and collagen. Our results open new lines of research on the role of PREP and α2M* in MS, aiming to relate them to the diagnosis and prognosis of this devastating disease.
Collapse
Affiliation(s)
- Jofre Tenorio-Laranga
- Division of Pharmacology and Toxicology, University of Helsinki, Viikinkaari 5E, 00014 Finland
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
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: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
29
|
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
|
30
|
Wang L, Chopp M, Szalad A, Liu Z, Lu M, Zhang L, Zhang J, Zhang RL, Morris D, Zhang ZG. Thymosin β4 promotes the recovery of peripheral neuropathy in type II diabetic mice. Neurobiol Dis 2012; 48:546-55. [PMID: 22922221 DOI: 10.1016/j.nbd.2012.08.002] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2012] [Revised: 07/20/2012] [Accepted: 08/02/2012] [Indexed: 01/10/2023] Open
Abstract
Peripheral neuropathy is one of the most common complications of diabetes mellitus. Using a mouse model of diabetic peripheral neuropathy, we tested the hypothesis that thymosin β4 (Tβ4) ameliorates diabetes-induced neurovascular dysfunction in the sciatic nerve and promotes recovery of neurological function from diabetic peripheral neuropathy. Tβ4 treatment of diabetic mice increased functional vascular density and regional blood flow in the sciatic nerve, and improved nerve function. Tβ4 upregulated angiopoietin-1 (Ang1) expression, but suppressed Ang2 expression in endothelial and Schwann cells in the diabetic sciatic nerve. In vitro, incubation of Human Umbilical Vein Endothelial Cells (HUVECs) with Tβ4 under high glucose condition completely abolished high glucose-downregulated Ang1 expression and high glucose-reduced capillary-like tube formation. Moreover, incubation of HUVECs under high glucose with conditioned medium collected from Human Schwann Cells (HSCs) treated with Tβ4 significantly reversed high glucose-decreased capillary-like tube formation. PI3K/Akt signaling pathway is involved in Tβ4-regulated Ang1 expression on endothelial and Schwann cells. These data indicate that Tβ4 likely acts on endothelial cells and Schwann cells to preserve and/or restore vascular function in the sciatic nerve which facilitates improvement of peripheral nerve function under diabetic neuropathy. Thus, Tβ4 has potential for the treatment of diabetic peripheral neuropathy.
Collapse
Affiliation(s)
- Lei Wang
- Department of Neurology, Henry Ford Hospital, 2799 W. Grand Boulevard, Detroit, MI 48202, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Xiong Y, Zhang Y, Mahmood A, Meng Y, Zhang ZG, Morris DC, Chopp M. Neuroprotective and neurorestorative effects of thymosin β4 treatment initiated 6 hours after traumatic brain injury in rats. J Neurosurg 2012; 116:1081-92. [PMID: 22324420 PMCID: PMC3392183 DOI: 10.3171/2012.1.jns111729] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
OBJECT Thymosin β4 (Tβ4) is a regenerative multifunctional peptide. The aim of this study was to test the hypothesis that Tβ4 treatment initiated 6 hours postinjury reduces brain damage and improves functional recovery in rats subjected to traumatic brain injury (TBI). METHODS Traumatic brain injury was induced by controlled cortical impact over the left parietal cortex in young adult male Wistar rats. The rats were randomly divided into the following groups: 1) saline group (n = 7); 2) 6 mg/kg Tβ4 group (n = 8); and 3) 30 mg/kg Tβ4 group (n = 8). Thymosin β4 or saline was administered intraperitoneally starting at 6 hours postinjury and again at 24 and 48 hours. An additional group of 6 animals underwent surgery without TBI (sham-injury group). Sensorimotor function and spatial learning were assessed using the modified Neurological Severity Score and the Morris water maze test, respectively. Animals were euthanized 35 days after injury, and brain sections were processed to assess lesion volume, hippocampal cell loss, cell proliferation, and neurogenesis after Tβ4 treatment. RESULTS Compared with saline administration, Tβ4 treatment initiated 6 hours postinjury significantly improved sensorimotor functional recovery and spatial learning, reduced cortical lesion volume and hippocampal cell loss, and enhanced cell proliferation and neurogenesis in the injured hippocampus. The high dose of Tβ4 showed better beneficial effects compared with the low-dose treatment. CONCLUSIONS Thymosin β4 treatment initiated 6 hours postinjury provides both neuroprotection and neurorestoration after TBI, indicating that Tβ4 has promising therapeutic potential in patients with TBI. These data warrant further investigation of the optimal dose and therapeutic window of Tβ4 treatment for TBI and the associated underlying mechanisms.
Collapse
Affiliation(s)
- Ye Xiong
- Department of Neurosurgery, Henry Ford Health System, Detroit, Michigan 48202, USA.
| | | | | | | | | | | | | |
Collapse
|
32
|
Lui KO, Bu L, Li RA, Chan CW. Pluripotent stem cell-based heart regeneration: From the developmental and immunological perspectives. ACTA ACUST UNITED AC 2012; 96:98-108. [DOI: 10.1002/bdrc.21004] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
33
|
Wnt1/βcatenin injury response activates the epicardium and cardiac fibroblasts to promote cardiac repair. EMBO J 2011; 31:429-42. [PMID: 22085926 PMCID: PMC3261567 DOI: 10.1038/emboj.2011.418] [Citation(s) in RCA: 255] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2011] [Accepted: 10/24/2011] [Indexed: 12/18/2022] Open
Abstract
Wnts are required for cardiogenesis but the role of specific Wnts in cardiac repair remains unknown. In this report, we show that a dynamic Wnt1/βcatenin injury response activates the epicardium and cardiac fibroblasts to promote cardiac repair. Acute ischaemic cardiac injury upregulates Wnt1 that is initially expressed in the epicardium and subsequently by cardiac fibroblasts in the region of injury. Following cardiac injury, the epicardium is activated organ-wide in a Wnt-dependent manner, expands, undergoes epithelial-mesenchymal transition (EMT) to generate cardiac fibroblasts, which localize in the subepicardial space. The injured regions in the heart are Wnt responsive as well and Wnt1 induces cardiac fibroblasts to proliferate and express pro-fibrotic genes. Disruption of downstream Wnt signalling in epicardial cells decreases epicardial expansion, EMT and leads to impaired cardiac function and ventricular dilatation after cardiac injury. Furthermore, disruption of Wnt/βcatenin signalling in cardiac fibroblasts impairs wound healing and decreases cardiac performance as well. These findings reveal that a pro-fibrotic Wnt1/βcatenin injury response is critically required for preserving cardiac function after acute ischaemic cardiac injury.
Collapse
|
34
|
Rupp H, Rupp TP, Alter P, Jung N, Pankuweit S, Maisch B. Intrapericardial procedures for cardiac regeneration by stem cells: need for minimal invasive access (AttachLifter) to the normal pericardial cavity. Herz 2011; 35:458-65. [PMID: 20941468 DOI: 10.1007/s00059-010-3382-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
In view of the only modest functional and anatomical improvements achieved by bone marrow-derived cell transplantation in patients with heart disease, the question was addressed whether the intracoronary, transcoronary-venous, and intramyocardial delivery routes are adequate. It is hypothesized that an intrapericardial delivery of stem cells or activators of resident cardiac stem cells increases therapeutic benefits. From such an intrapericardial depot, cells or modulating factors, such as thymosin β4 or Ac-SDKP, are expected to reach the myocardium with sustained kinetics. Novel tools which provide access to the pericardial space even in the absence of pericardial effusion are, therefore, described. When the pericardium becomes attached to the suction head (monitored by an increase in negative pressure), the pericardium is lifted from the epicardium ("AttachLifter"). The opening of the suction head ("Attacher") is narrowed by flexible clamps which grab the tissue and improve the vacuum seal in the case of uneven tissue. A ridge, i.e.,"needle guidance", on the suction head excludes injury to the epicardium, whereby the pericardium is punctured by a needle which resides outside the suction head. A fiberscope can be used to inspect the pericardium prior to puncture. Based on these procedures, the role of the pericardial space and the presence of pericardial effusion in cardiac regeneration can be assessed.
Collapse
Affiliation(s)
- H Rupp
- Department of Internal Medicine - Cardiology, Experimental Cardiology Laboratory, Philipps University of Marburg, Marburg, Deutschland.
| | | | | | | | | | | |
Collapse
|
35
|
Freeman KW, Bowman BR, Zetter BR. Regenerative protein thymosin beta-4 is a novel regulator of purinergic signaling. FASEB J 2011; 25:907-15. [PMID: 21106936 DOI: 10.1096/fj.10-169417] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2023]
Abstract
By an unknown mechanism, β-thymosins are extracellular modulators of angiogenesis, inflammation, wound healing, and development. We were interested in identifying β-thymosin interactors and determining their importance in β-thymosins signaling in human vein endothelial cells (HUVECs). We performed pulldown experiments with biotinylated thymosin β-4 (Tβ4) in comparison to neutravidin beads alone and used mass spectrometric analysis to identify differentially interacting proteins. By this method, we identified F1-F0 ATP synthase, a known target of antiangiogenic angiostatin. By surface plasmon resonance, we determined for Tβ4 binding to the β subunit of ATP synthase a K(D) of 12 nM. Blocking antibodies and antagonists (oligomycin, IC(50) ∼1.8 μM; piceatannol, IC(50) ∼1.05 μM; and angiostatin, IC(50) ∼2.9 μg/ml) of ATP synthase inhibited the Tβ4-induced increase in cell surface ATP levels, as measured by luciferase assay, and the Tβ4-induced increase in HUVEC migration, as measured by transwell migration assay. Silencing of the ATP-responsive purinergic receptor P2X4 with siRNA also blocked Tβ4-induced HUVEC migration in a transwell assay. Furthermore, in silico we identified common amphiphilic α-helical structural similarities between β-thymosins and the inhibitory factor 1 (IF1), an inhibitor of ATP synthase hydrolysis. In summary, we have identified an extracellular signaling pathway where Tβ4 increases cell surface ATP levels via ATP synthase and have shown further that ATP-responsive P2X4 receptor is required for Tβ4-induced HUVEC migration.
Collapse
Affiliation(s)
- Kevin W Freeman
- Vascular Biology Program and Department of Surgery, Children's Hospital, Boston, MA 02115, USA
| | | | | |
Collapse
|
36
|
The epicardium in cardiac repair: From the stem cell view. Pharmacol Ther 2011; 129:82-96. [DOI: 10.1016/j.pharmthera.2010.09.002] [Citation(s) in RCA: 73] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2010] [Accepted: 09/09/2010] [Indexed: 12/12/2022]
|
37
|
Affiliation(s)
- Jonathan A Epstein
- Department of Cell and Developmental Biology and the Cardiovascular Institute, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA.
| |
Collapse
|
38
|
Identification of Thymosin β4 as an effector of Hand1-mediated vascular development. Nat Commun 2010; 1:46. [PMID: 20975697 PMCID: PMC2963826 DOI: 10.1038/ncomms1041] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2009] [Accepted: 06/29/2010] [Indexed: 01/25/2023] Open
Abstract
The bHLH transcription factor Hand1 (Heart and neural crest-derived transcript-1) has a fundamental role in cardiovascular development; however, the molecular mechanisms have not been elucidated. In this paper we identify Thymosin β4 (Tβ4/Tmsb4x), which encodes an actin monomer-binding protein implicated in cell migration and angiogenesis, as a direct target of Hand1. We demonstrate that Hand1 binds an upstream regulatory region proximal to the promoter of Tβ4 at consensus Thing1 and E-Box sites and identify both activation and repression of Tβ4 by Hand1, through direct binding within either non-canonical or canonical E-boxes, providing new insight into gene regulation by bHLH transcription factors. Hand1-mediated activation of Tβ4 is essential for yolk sac vasculogenesis and embryonic survival, and administration of synthetic TB4 partially rescues yolk sac capillary plexus formation in Hand1-null embryos. Thus, we identify an in vivo downstream target of Hand1 and reveal impaired yolk sac vasculogenesis as a primary cause of early embryonic lethality following loss of this critical bHLH factor. The Hand1 transcription factor plays a central role in cardiovascular development. Here the authors demonstrate that Hand1 regulates thymosin β4 and that the delivery of synthetic thymosin β4 can rescue some of the vascular defects in Hand1 null mouse embryos.
Collapse
|
39
|
Bollini S, Smart N, Riley PR. Resident cardiac progenitor cells: at the heart of regeneration. J Mol Cell Cardiol 2010; 50:296-303. [PMID: 20643135 DOI: 10.1016/j.yjmcc.2010.07.006] [Citation(s) in RCA: 96] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2010] [Revised: 07/02/2010] [Accepted: 07/08/2010] [Indexed: 10/19/2022]
Abstract
Stem cell therapy has recently emerged as an innovative strategy over conventional cardiovascular treatments to restore cardiac function in patients affected by ischemic heart disease. Various stem cell populations have been tested and their potential for cardiac repair has been analyzed. Embryonic stem cells retain the greatest differentiation potential, but concerns persist with regard to their immunogenic and teratogenic effects. Although adult somatic stem cells are not tumourigenic and easier to use in an autologous setting, they exist in small numbers and possess reduced differentiation potential. Traditionally the heart was considered to be a post-mitotic organ; however, this dogma has recently been challenged with the identification of a reservoir of resident stem cells, defined as cardiac progenitor cells (CPCs). These endogenous progenitors may represent the best candidates for cardiovascular cell therapy, as they are tissue-specific, often pre-committed to a cardiac fate, and display a greater propensity to differentiate towards cardiovascular lineages. This review will focus on current research into the biology of CPCs and their regenerative potential. This article is part of a special issue entitled, "Cardiovascular Stem Cells Revisited".
Collapse
Affiliation(s)
- Sveva Bollini
- Molecular Medicine Unit, University College London-Institute of Child Health, London, UK
| | | | | |
Collapse
|
40
|
Hinkel R, Bock-Marquette I, Hatzopoulos AK, Hazopoulos AK, Kupatt C. Thymosin beta4: a key factor for protective effects of eEPCs in acute and chronic ischemia. Ann N Y Acad Sci 2010; 1194:105-11. [PMID: 20536456 DOI: 10.1111/j.1749-6632.2010.05489.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Acute myocardial infarction is still one of the leading causes of death in the industrial nations. Even after successful revascularization, myocardial ischemia results in a loss of cardiomyocytes and scar formation. Embryonic EPCs (eEPCs), retroinfused into the ischemic region of the pig heart, provided rapid paracrine benefit to acute and chronic ischemia in a PI-3K/Akt-dependent manner. In a model of acute myocardial ischemia, infarct size and loss of regional myocardial function decreased after eEPC application, unless cell pre-treatment with thymosin beta4 shRNA was performed. Thymosin beta4 peptide retroinfusion mimicked the eEPC-derived improvement of infarct size and myocardial function. In chronic ischemia (rabbit model), eEPCs retroinfused into the ischemic hindlimb enhanced capillary density, collateral growth, and perfusion. Therapeutic neovascularization was absent when thymosin beta4 shRNA was introduced into eEPCs before application. In conclusion, eEPCs are capable of acute and chronic ischemia protection in a thymosin beta4 dependent manner.
Collapse
Affiliation(s)
- Rabea Hinkel
- Internal Medicine I, Klinikum Grosshadern, Ludwig Maximilians University, Munich, Germany
| | | | | | | | | |
Collapse
|
41
|
Revealing new mouse epicardial cell markers through transcriptomics. PLoS One 2010; 5:e11429. [PMID: 20596535 PMCID: PMC2893200 DOI: 10.1371/journal.pone.0011429] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2009] [Accepted: 05/24/2010] [Indexed: 12/29/2022] Open
Abstract
Background The epicardium has key functions during myocardial development, by contributing to the formation of coronary endothelial and smooth muscle cells, cardiac fibroblasts, and potentially cardiomyocytes. The epicardium plays a morphogenetic role by emitting signals to promote and maintain cardiomyocyte proliferation. In a regenerative context, the adult epicardium might comprise a progenitor cell population that can be induced to contribute to cardiac repair. Although some genes involved in epicardial function have been identified, a detailed molecular profile of epicardial gene expression has not been available. Methodology Using laser capture microscopy, we isolated the epicardial layer from the adult murine heart before or after cardiac infarction in wildtype mice and mice expressing a transgenic IGF-1 propeptide (mIGF-1) that enhances cardiac repair, and analyzed the transcription profile using DNA microarrays. Principal Findings Expression of epithelial genes such as basonuclin, dermokine, and glycoprotein M6A are highly enriched in the epicardial layer, which maintains expression of selected embryonic genes involved in epicardial development in mIGF-1 transgenic hearts. After myocardial infarct, a subset of differentially expressed genes are down-regulated in the epicardium representing an epicardium-specific signature that responds to injury. Conclusion This study presents the description of the murine epicardial transcriptome obtained from snap frozen tissues, providing essential information for further analysis of this important cardiac cell layer.
Collapse
|
42
|
Xiong Y, Mahmood A, Meng Y, Zhang Y, Zhang ZG, Morris DC, Chopp M. Treatment of traumatic brain injury with thymosin β₄ in rats. J Neurosurg 2010; 114:102-15. [PMID: 20486893 DOI: 10.3171/2010.4.jns10118] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
OBJECT This study was designed to investigate the efficacy of delayed thymosin β(4) (Tβ(4)) treatment of traumatic brain injury (TBI) in rats. METHODS Young adult male Wistar rats were divided into the following groups: 1) sham group (6 rats); 2) TBI + saline group (9 rats); 3) and TBI + Tβ(4) group (10 rats). Traumatic brain injury was induced by controlled cortical impact over the left parietal cortex. Thymosin β(4) (6 mg/kg) or saline was administered intraperitoneally starting at Day 1 and then every 3 days for an additional 4 doses. Neurological function was assessed using a modified neurological severity score (mNSS), foot fault, and Morris water maze tests. Animals were killed 35 days after injury, and brain sections were stained for immunohistochemistry to assess angiogenesis, neurogenesis, and oligodendrogenesis after Tβ(4) treatment. RESULTS Compared with the saline treatment, delayed Tβ(4) treatment did not affect lesion volume but significantly reduced hippocampal cell loss, enhanced angiogenesis and neurogenesis in the injured cortex and hippocampus, increased oligodendrogenesis in the CA3 region, and significantly improved sensorimotor functional recovery and spatial learning. CONCLUSIONS These data for the first time demonstrate that delayed administration of Tβ(4) significantly improves histological and functional outcomes in rats with TBI, indicating that Tβ(4) has considerable therapeutic potential for patients with TBI.
Collapse
Affiliation(s)
- Ye Xiong
- Department of Neurosurgery, Henry Ford Health System, Detroit, Michigan 48202, USA.
| | | | | | | | | | | | | |
Collapse
|
43
|
Hwang H, Kloner RA. Improving regenerating potential of the heart after myocardial infarction: factor-based approach. Life Sci 2010; 86:461-72. [PMID: 20093126 DOI: 10.1016/j.lfs.2010.01.004] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2009] [Revised: 12/28/2009] [Accepted: 01/11/2010] [Indexed: 01/09/2023]
Abstract
The emerging evidence that the heart has the potential to regenerate, albeit not ideally, has stimulated considerable interest in the field of cardiac regenerative medicine. Several lines of research demonstrated that factor-based therapy is feasible and effective, whether it is used independently or as an adjunct to cell therapy. The ultimate goal of the factor-based approach is to improve the regenerating potential of the heart as a means to treat patients with cardiovascular disease. This article reviews recent approaches involving factor-based therapy for cardiac repair and regeneration including some of the advantages of this type of therapy as well as some of the hurdles that must be overcome before this therapeutic approach becomes a standard part of clinical medicine.
Collapse
Affiliation(s)
- Hyosook Hwang
- Heart Institute, Good Samaritan Hospital, Los Angeles, CA 90017, United States
| | | |
Collapse
|
44
|
Zhang J, Zhang ZG, Morris D, Li Y, Roberts C, Elias SB, Chopp M. Neurological functional recovery after thymosin beta4 treatment in mice with experimental auto encephalomyelitis. Neuroscience 2009; 164:1887-93. [PMID: 19782721 DOI: 10.1016/j.neuroscience.2009.09.054] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2009] [Revised: 09/21/2009] [Accepted: 09/21/2009] [Indexed: 02/07/2023]
Abstract
In the present study, we hypothesized that thymosin beta 4 (Tbeta4) is a potential therapy of multiple sclerosis (MS). To test this hypothesis, SJL/J mice (n=21) were subjected to experimental autoimmune encephalomyelitis (EAE), an animal model of MS. EAE mice were treated with saline or Tbeta4 (6 mg/kg, n=10) every 3 days starting on the day of myelin proteolipid protein (PLP) immunization for total five doses. Neurological function, inflammatory infiltration, oligodendrocyte progenitor cells (OPCs) and mature oligodendrocytes were measured in the brain of EAE mice. Double immunohistochemical staining was used to detect proliferation and differentiation of OPCs. Tbeta4 was used to treat N20.1 cells (premature oligodendrocyte cell line) in vitro, and proliferation of N20.1 cells was measured by bromodeoxyuridine (BrdU) immunostaining. Tbeta4 treatment improved functional recovery after EAE. Inflammatory infiltrates were significantly reduced in the Tbeta4 treatment group compared to the saline groups (3.6+/-0.3/slide vs 5+/-0.5/slide, P<0.05). NG2(+) OPCs (447.7+/-41.9 vs 195.2+/-31/mm(2) in subventricular zone (SVZ), 75.1+/-4.7 vs 41.7+/-3.2/mm(2) in white matter), CNPase(+) mature oligodendrocytes (267.5+/-10.3 vs 141.4+/-22.9/mm(2)), BrdU(+) with NG2(+) OPCs (32.9+/-3.7 vs 17.9+/-3.6/mm(2)), BrdU(+) with CNPase(+) mature oligodendrocytes (18.2+/-1.7 vs 10.7+/-2.2/mm(2)) were significantly increased in the Tbeta4 treated mice compared to those of saline controls (P<0.05). These data indicate that Tbeta4 treatment improved functional recovery after EAE, possibly, via reducing inflammatory infiltrates, and stimulating oligodendrogenesis.
Collapse
Affiliation(s)
- J Zhang
- Department of Neurology, Henry Ford Health System, Detroit, MI 48202, USA
| | | | | | | | | | | | | |
Collapse
|
45
|
Thymosin beta4 induces endothelial progenitor cell migration via PI3K/Akt/eNOS signal transduction pathway. J Cardiovasc Pharmacol 2009; 53:209-14. [PMID: 19247195 DOI: 10.1097/fjc.0b013e318199f326] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Thymosin beta4, a G-actin-sequestering peptide, has been shown to play an important role in cell migration. However, little is known about the effect of thymosin beta4 on circulating endothelial progenitor cell (EPC) directional migration, which is essential for EPC-mediated reendothelialization and neovascularization. In our study, using a transwell migration assay, we showed that thymosin beta4 induced EPC migration in a concentration-dependent manner. Western blot analysis revealed that treatment of EPCs with thymosin beta4 resulted in a time and concentration-dependent phosphorylation of Akt, endothelial nitric oxide synthase (eNOS), and extracellular signal-regulated kinase (ERK)1/2. Functional analysis showed that thymosin beta4-induced EPC migration was blocked by phosphatidylinositol 3-kinase inhibitors (LY294002 or wortmannin) or eNOS inhibitor (Nomega-nitro-L-arginine methyl ester) but was not significantly attenuated by mitogen-activated protein kinase (MAPK)/ERK inhibitor (PD98059). These findings suggest that thymosin beta4 stimulates EPC directional migration via phosphatidylinositol 3-kinase/Akt/eNOS, rather than via MAPK/ERK signal transduction pathway.
Collapse
|
46
|
Smart N, Riley PR. Derivation of epicardium-derived progenitor cells (EPDCs) from adult epicardium. CURRENT PROTOCOLS IN STEM CELL BIOLOGY 2009; Chapter 2:Unit2C.2. [PMID: 19235142 DOI: 10.1002/9780470151808.sc02c02s8] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The epicardium has, like the other cell lineages of the terminally differentiated adult heart, long been regarded as quiescent, incapable of migration or differentiation. In contrast, the embryonic epicardium possesses an innate ability to proliferate, migrate, and differentiate into a number of mature cardiovascular cell types, including vascular smooth muscle cells, fibroblasts, cardiomyocytes, and, arguably, some endothelial cells. In recapitulating its essential developmental role, we recognized the ability of the actin-binding peptide thymosin beta4 (Tbeta4) to induce epicardium-derived progenitor cell (EPDC) migration from adult heart and noted the derivation of cell types originating from embryonic epicardium. This protocol provides a means of enabling adult EPDC outgrowth and culture. We establish a model system in which to study the ability of factors to influence the migration of vascular precursors and their differentiation and to move towards screening of small molecules ex vivo prior to clinical trials of therapeutic cardiac repair.
Collapse
Affiliation(s)
- Nicola Smart
- UCL Institute of Child Health, London, United Kingdom
| | | |
Collapse
|
47
|
Abstract
Human embryonic stem (HES) cells are pluripotent and give rise to any cell lineage. More specifically, how the first embryonic lineage (i.e., cardiac lineage) is acquired remains in many aspects questionable. Herein, we summarize the protocols that have been used to direct the fate of HES cells toward the cardiomyocytic lineage. We further discuss the regulation of transcriptional pathways underlying this process of differentiation. Finally, we propose perspectives of this research in the near future.
Collapse
|
48
|
Aitsebaomo J, Portbury AL, Schisler JC, Patterson C. Brothers and sisters: molecular insights into arterial-venous heterogeneity. Circ Res 2008; 103:929-39. [PMID: 18948631 DOI: 10.1161/circresaha.108.184937] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
The molecular differences between arteries and veins are genetically predetermined and are evident even before the first embryonic heart beat. Although ephrinB2 and EphB4 are expressed in cells that will ultimately differentiate into arteries and veins, respectively, many other genes have been shown to play a significant role in cell fate determination. The expression patterns of ephrinB2 and EphB4 are restricted to arterial-venous boundaries, and Eph/ephrin signaling provides repulsive cues at arterial-venous boundaries that are thought to prevent intermixing of arterial- and venous-fated cells. However, the maintenance of arterial-venous fate is susceptible to some degree of plasticity. Thus, in response to signals from the ambient microenvironment and shear stress, there is flow-mediated intercalation of the arteries and veins that ultimately leads to the formation of a functional, closed-loop circulation. In addition, cells in the blood vessels of each organ undergo epigenetic, morphological, and functional adaptive changes that are specific to the proximate function of their cognate organ(s). These adaptive changes result in an interorgan and intraorgan vessel heterogeneity that manifest clinically in a disparate response of different organs to identical risk factors and injury in the same animal. In this review, we focus on the molecular and physiological factors influencing arterial-venous heterogeneity between and within different organ(s). We explore arterial-venous differences in selected organs, as well as their respective endothelial cell architectural organization that results in their inter- and intraorgan heterogeneity.
Collapse
Affiliation(s)
- Julius Aitsebaomo
- Division of Cardiology and Carolina Cardiovascular Biology Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-7126, USA
| | | | | | | |
Collapse
|
49
|
Abstract
Nuclear magnetic resonance (NMR) spectroscopy is a versatile biophysical technique with wide applicability in drug discovery research, particularly for the detection and characterization of molecular interactions. This review highlights in a comprehensive manner the aspects of biomolecular NMR which are most beneficial for pharmaceutical research and presents them as contributions to the different stages of a drug discovery program: target selection, assay development, lead generation and lead optimization. Emphasis is put on the concept of the particular NMR application, rather than on technical details, and on recent examples. Finally, an appendix of frequently asked questions is given.
Collapse
Affiliation(s)
- W Jahnke
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, Lichtstrasse, 4002, Basel, Switzerland.
| | - H Widmer
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, Lichtstrasse, 4002, Basel, Switzerland
| |
Collapse
|