1
|
Kiskin FN, Yang Y, Yang H, Zhang JZ. Cracking the code of the cardiovascular enigma: hPSC-derived endothelial cells unveil the secrets of endothelial dysfunction. J Mol Cell Cardiol 2024; 192:65-78. [PMID: 38761989 DOI: 10.1016/j.yjmcc.2024.05.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 05/08/2024] [Accepted: 05/10/2024] [Indexed: 05/20/2024]
Abstract
Endothelial dysfunction is a central contributor to the development of most cardiovascular diseases and is characterised by the reduced synthesis or bioavailability of the vasodilator nitric oxide together with other abnormalities such as inflammation, senescence, and oxidative stress. The use of patient-specific and genome-edited human pluripotent stem cell-derived endothelial cells (hPSC-ECs) has shed novel insights into the role of endothelial dysfunction in cardiovascular diseases with strong genetic components such as genetic cardiomyopathies and pulmonary arterial hypertension. However, their utility in studying complex multifactorial diseases such as atherosclerosis, metabolic syndrome and heart failure poses notable challenges. In this review, we provide an overview of the different methods used to generate and characterise hPSC-ECs before comprehensively assessing their effectiveness in cardiovascular disease modelling and high-throughput drug screening. Furthermore, we explore current obstacles that will need to be overcome to unleash the full potential of hPSC-ECs in facilitating patient-specific precision medicine. Addressing these challenges holds great promise in advancing our understanding of intricate cardiovascular diseases and in tailoring personalised therapeutic strategies.
Collapse
Affiliation(s)
- Fedir N Kiskin
- Institute of Neurological and Psychiatric Disorders, Shenzhen Bay Laboratory, Shenzhen 518132, China.
| | - Yuan Yang
- Institute of Neurological and Psychiatric Disorders, Shenzhen Bay Laboratory, Shenzhen 518132, China.
| | - Hao Yang
- Institute of Neurological and Psychiatric Disorders, Shenzhen Bay Laboratory, Shenzhen 518132, China.
| | - Joe Z Zhang
- Institute of Neurological and Psychiatric Disorders, Shenzhen Bay Laboratory, Shenzhen 518132, China.
| |
Collapse
|
2
|
Li RG, Li X, Morikawa Y, Grisanti-Canozo FJ, Meng F, Tsai CR, Zhao Y, Liu L, Kim J, Xie B, Klysik E, Liu S, Samee MAH, Martin JF. YAP induces a neonatal-like pro-renewal niche in the adult heart. NATURE CARDIOVASCULAR RESEARCH 2024; 3:283-300. [PMID: 38510108 PMCID: PMC10954255 DOI: 10.1038/s44161-024-00428-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Accepted: 01/12/2024] [Indexed: 03/22/2024]
Abstract
After myocardial infarction (MI), mammalian hearts do not regenerate, and the microenvironment is disrupted. Hippo signaling loss of function with activation of transcriptional co-factor YAP induces heart renewal and rebuilds the post-MI microenvironment. In this study, we investigated adult renewal-competent mouse hearts expressing an active version of YAP, called YAP5SA, in cardiomyocytes (CMs). Spatial transcriptomics and single-cell RNA sequencing revealed a conserved, renewal-competent CM cell state called adult (a)CM2 with high YAP activity. aCM2 co-localized with cardiac fibroblasts (CFs) expressing complement pathway component C3 and macrophages (MPs) expressing C3ar1 receptor to form a cellular triad in YAP5SA hearts and renewal-competent neonatal hearts. Although aCM2 was detected in adult mouse and human hearts, the cellular triad failed to co-localize in these non-renewing hearts. C3 and C3ar1 loss-of-function experiments indicated that C3a signaling between MPs and CFs was required to assemble the pro-renewal aCM2, C3+ CF and C3ar1+ MP cellular triad.
Collapse
Affiliation(s)
- Rich Gang Li
- Cardiomyocyte Renewal Laboratory, Texas Heart Institute, Houston, TX, USA
- McGill Gene Editing Laboratory, Texas Heart Institute, Houston, TX, USA
- These authors contributed equally: Rich Gang Li, Xiao Li
| | - Xiao Li
- McGill Gene Editing Laboratory, Texas Heart Institute, Houston, TX, USA
- These authors contributed equally: Rich Gang Li, Xiao Li
| | - Yuka Morikawa
- Cardiomyocyte Renewal Laboratory, Texas Heart Institute, Houston, TX, USA
| | - Francisco J. Grisanti-Canozo
- Cardiomyocyte Renewal Laboratory, Texas Heart Institute, Houston, TX, USA
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, USA
| | - Fansen Meng
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, USA
| | - Chang-Ru Tsai
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, USA
| | - Yi Zhao
- McGill Gene Editing Laboratory, Texas Heart Institute, Houston, TX, USA
| | - Lin Liu
- McGill Gene Editing Laboratory, Texas Heart Institute, Houston, TX, USA
| | - Jong Kim
- Cardiomyocyte Renewal Laboratory, Texas Heart Institute, Houston, TX, USA
| | - Bing Xie
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, USA
| | - Elzbieta Klysik
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, USA
| | - Shijie Liu
- Cardiomyocyte Renewal Laboratory, Texas Heart Institute, Houston, TX, USA
| | - Md Abul Hassan Samee
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, USA
| | - James F. Martin
- Cardiomyocyte Renewal Laboratory, Texas Heart Institute, Houston, TX, USA
- McGill Gene Editing Laboratory, Texas Heart Institute, Houston, TX, USA
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, USA
- Center for Organ Repair and Renewal, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
3
|
Górska A, Mazur AJ. Integrin-linked kinase (ILK): the known vs. the unknown and perspectives. Cell Mol Life Sci 2022; 79:100. [PMID: 35089438 PMCID: PMC8799556 DOI: 10.1007/s00018-021-04104-1] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 11/29/2021] [Accepted: 12/17/2021] [Indexed: 02/08/2023]
Abstract
Integrin-linked kinase (ILK) is a multifunctional molecular actor in cell-matrix interactions, cell adhesion, and anchorage-dependent cell growth. It combines functions of a signal transductor and a scaffold protein through its interaction with integrins, then facilitating further protein recruitment within the ILK-PINCH-Parvin complex. ILK is involved in crucial cellular processes including proliferation, survival, differentiation, migration, invasion, and angiogenesis, which reflects on systemic changes in the kidney, heart, muscle, skin, and vascular system, also during the embryonal development. Dysfunction of ILK underlies the pathogenesis of various diseases, including the pro-oncogenic activity in tumorigenesis. ILK localizes mostly to the cell membrane and remains an important component of focal adhesion. We do know much about ILK but a lot still remains either uncovered or unclear. Although it was initially classified as a serine/threonine-protein kinase, its catalytical activity is now questioned due to structural and functional issues, leaving the exact molecular mechanism of signal transduction by ILK unsolved. While it is known that the three isoforms of ILK vary in length, the presence of crucial domains, and modification sites, most of the research tends to focus on the main isoform of this protein while the issue of functional differences of ILK2 and ILK3 still awaits clarification. The activity of ILK is regulated on the transcriptional, protein, and post-transcriptional levels. The crucial role of phosphorylation and ubiquitylation has been investigated, but the functions of the vast majority of modifications are still unknown. In the light of all those open issues, here we present an extensive literature survey covering a wide spectrum of latest findings as well as a past-to-present view on controversies regarding ILK, finishing with pointing out some open questions to be resolved by further research.
Collapse
Affiliation(s)
- Agata Górska
- Department of Cell Pathology, Faculty of Biotechnology, University of Wroclaw, ul. Joliot-Curie 14a, 50-383, Wrocław, Poland.
| | - Antonina Joanna Mazur
- Department of Cell Pathology, Faculty of Biotechnology, University of Wroclaw, ul. Joliot-Curie 14a, 50-383, Wrocław, Poland.
| |
Collapse
|
4
|
Su L, Kong X, Loo S, Gao Y, Liu B, Su X, Dalan R, Ma J, Ye L. Thymosin beta-4 improves endothelial function and reparative potency of diabetic endothelial cells differentiated from patient induced pluripotent stem cells. Stem Cell Res Ther 2022; 13:13. [PMID: 35012642 PMCID: PMC8751378 DOI: 10.1186/s13287-021-02687-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 12/17/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Prior studies show that signature phenotypes of diabetic human induced pluripotent stem cells derived endothelial cells (dia-hiPSC-ECs) are disrupted glycine homeostasis, increased senescence, impaired mitochondrial function and angiogenic potential as compared with healthy hiPSC-ECs. In the current study, we aimed to assess the role of thymosin β-4 (Tb-4) on endothelial function using dia-hiPSC-ECs as disease model of endothelial dysfunction. METHODS AND RESULTS Using dia-hiPSC-ECs as models of endothelial dysfunction, we determined the effect of Tb-4 on cell proliferation, senescence, cyto-protection, protein expression of intercellular adhesion molecule-1 (ICAM-1), secretion of endothelin-1 and MMP-1, mitochondrial membrane potential, and cyto-protection in vitro and angiogenic potential for treatment of ischemic limb disease in a mouse model of type 2 diabetes mellitus (T2DM) in vivo. We found that 600 ng/mL Tb4 significantly up-regulated AKT activity and Bcl-XL protein expression, enhanced dia-hiPSC-EC viability and proliferation, limited senescence, reduced endothelin-1 and MMP-1 secretion, and improved reparative potency of dia-hiPSC-ECs for treatment of ischemic limb disease in mice with T2DM. However, Tb4 had no effect on improving mitochondrial membrane potential and glycine homeostasis and reducing intercellular adhesion molecule-1 protein expression in dia-hiPSC-ECs. CONCLUSIONS Tb-4 improves endothelial dysfunction through enhancing hiPSC-EC viability, reducing senescence and endothelin-1 production, and improves angiogenic potency in diabetes.
Collapse
Affiliation(s)
- Liping Su
- National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore, 169609 Singapore
| | - Xiaocen Kong
- Department of Endocrinology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210029 China
| | - Szejie Loo
- National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore, 169609 Singapore
| | - Yu Gao
- Department of Cardiology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127 China
| | - Bingli Liu
- Department of Endocrinology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210029 China
| | - Xiaofei Su
- Department of Endocrinology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210029 China
| | - Rinkoo Dalan
- Department of Endocrinology, Tan Tock Seng Hospital, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Jianhua Ma
- Department of Endocrinology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210029 China
| | - Lei Ye
- National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore, 169609 Singapore
| |
Collapse
|
5
|
Scheller I, Beck S, Göb V, Gross C, Neagoe RAI, Aurbach K, Bender M, Stegner D, Nagy Z, Nieswandt B. Thymosin β4 is essential for thrombus formation by controlling the G-actin/F-actin equilibrium in platelets. Haematologica 2021; 107:2846-2858. [PMID: 34348450 PMCID: PMC9713564 DOI: 10.3324/haematol.2021.278537] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Indexed: 12/14/2022] Open
Abstract
Coordinated rearrangements of the actin cytoskeleton are pivotal for platelet biogenesis from megakaryocytes but also orchestrate key functions of peripheral platelets in hemostasis and thrombosis, such as granule release, the formation of filopodia and lamellipodia, or clot retraction. Along with profilin (Pfn) 1, thymosin β4 (encoded by Tmsb4x) is one of the two main G-actin-sequestering proteins within cells of higher eukaryotes, and its intracellular concentration is particularly high in cells that rapidly respond to external signals by increased motility, such as platelets. Here, we analyzed constitutive Tmsb4x knockout (KO) mice to investigate the functional role of the protein in platelet production and function. Thymosin β4 deficiency resulted in a macrothrombocytopenia with only mildly increased platelet volume and an unaltered platelet life span. Megakaryocyte numbers in the bone marrow and spleen were unaltered, however, Tmsb4x KO megakaryocytes showed defective proplatelet formation in vitro and in vivo. Thymosin β4-deficient platelets displayed markedly decreased G-actin levels and concomitantly increased F-actin levels resulting in accelerated spreading on fibrinogen and clot retraction. Moreover, Tmsb4x KO platelets showed activation defects and an impaired immunoreceptor tyrosine-based activation motif (ITAM) signaling downstream of the activating collagen receptor glycoprotein VI. These defects translated into impaired aggregate formation under flow, protection from occlusive arterial thrombus formation in vivo and increased tail bleeding times. In summary, these findings point to a critical role of thymosin β4 for actin dynamics during platelet biogenesis, platelet activation downstream of glycoprotein VI and thrombus stability.
Collapse
Affiliation(s)
- Inga Scheller
- Institute of Experimental Biomedicine I, University Hospital, University of Würzburg, and Rudolf Virchow Center for Integrative and Translational BioImaging, University of Würzburg, Würzburg, Germany,*IS and SB contributed equally as co-first authors
| | - Sarah Beck
- Institute of Experimental Biomedicine I, University Hospital, University of Würzburg, and Rudolf Virchow Center for Integrative and Translational BioImaging, University of Würzburg, Würzburg, Germany,*IS and SB contributed equally as co-first authors
| | - Vanessa Göb
- Institute of Experimental Biomedicine I, University Hospital, University of Würzburg, and Rudolf Virchow Center for Integrative and Translational BioImaging, University of Würzburg, Würzburg, Germany
| | - Carina Gross
- Institute of Experimental Biomedicine I, University Hospital, University of Würzburg, and Rudolf Virchow Center for Integrative and Translational BioImaging, University of Würzburg, Würzburg, Germany
| | - Raluca A. I. Neagoe
- Institute of Experimental Biomedicine I, University Hospital, University of Würzburg, and Rudolf Virchow Center for Integrative and Translational BioImaging, University of Würzburg, Würzburg, Germany,Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham, UK
| | - Katja Aurbach
- Institute of Experimental Biomedicine I, University Hospital, University of Würzburg, and Rudolf Virchow Center for Integrative and Translational BioImaging, University of Würzburg, Würzburg, Germany
| | - Markus Bender
- Institute of Experimental Biomedicine I, University Hospital, University of Würzburg, and Rudolf Virchow Center for Integrative and Translational BioImaging, University of Würzburg, Würzburg, Germany
| | - David Stegner
- Institute of Experimental Biomedicine I, University Hospital, University of Würzburg, and Rudolf Virchow Center for Integrative and Translational BioImaging, University of Würzburg, Würzburg, Germany
| | - Zoltan Nagy
- Institute of Experimental Biomedicine I, University Hospital, University of Würzburg, and Rudolf Virchow Center for Integrative and Translational BioImaging, University of Würzburg, Würzburg, Germany
| | - Bernhard Nieswandt
- Institute of Experimental Biomedicine I, University Hospital, University of Würzburg, and Rudolf Virchow Center for Integrative and Translational BioImaging, University of Würzburg, Würzburg, Germany,B. Nieswandt
| |
Collapse
|
6
|
Tan SH, Loo SJ, Gao Y, Tao ZH, Su LP, Wang CX, Zhang SL, Mu YH, Cui YH, Abdurrachim D, Wang WH, Lalic J, Lim KC, Bu J, Tan RS, Lee TH, Zhang J, Ye L. Thymosin β4 increases cardiac cell proliferation, cell engraftment, and the reparative potency of human induced-pluripotent stem cell-derived cardiomyocytes in a porcine model of acute myocardial infarction. Am J Cancer Res 2021; 11:7879-7895. [PMID: 34335970 PMCID: PMC8315077 DOI: 10.7150/thno.56757] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 06/14/2021] [Indexed: 12/12/2022] Open
Abstract
Rationale: Previous studies have shown that human embryonic stem cell-derived cardiomyocytes improved myocardial recovery when administered to infarcted pig and non-human primate hearts. However, the engraftment of intramyocardially delivered cells is poor and the effectiveness of clinically relevant doses of human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) in large animal models of myocardial injury remains unknown. Here, we determined whether thymosin β4 (Tb4) could improve the engraftment and reparative potency of transplanted hiPSC-CMs in a porcine model of myocardial infarction (MI). Methods: Tb4 was delivered from injected gelatin microspheres, which extended the duration of Tb4 administration for up to two weeks in vitro. After MI induction, pigs were randomly distributed into 4 treatment groups: the MI Group was injected with basal medium; the Tb4 Group received gelatin microspheres carrying Tb4; the CM Group was treated with 1.2 × 108 hiPSC-CMs; and the Tb4+CM Group received both the Tb4 microspheres and hiPSC-CMs. Myocardial recovery was assessed by cardiac magnetic resonance imaging (MRI), arrhythmogenesis was monitored with implanted loop recorders, and tumorigenesis was evaluated via whole-body MRI. Results: In vitro, 600 ng/mL of Tb4 protected cultured hiPSC-CMs from hypoxic damage by upregulating AKT activity and BcL-XL and promoted hiPSC-CM and hiPSC-EC proliferation. In infarcted pig hearts, hiPSC-CM transplantation alone had a minimal effect on myocardial recovery, but co-treatment with Tb4 significantly enhanced hiPSC-CM engraftment, induced vasculogenesis and the proliferation of cardiomyocytes and endothelial cells, improved left ventricular systolic function, and reduced infarct size. hiPSC-CM implantation did not increase incidence of ventricular arrhythmia and did not induce tumorigenesis in the immunosuppressed pigs. Conclusions: Co-treatment with Tb4-microspheres and hiPSC-CMs was safe and enhanced the reparative potency of hiPSC-CMs for myocardial repair in a large-animal model of MI.
Collapse
|
7
|
Bjørklund G, Dadar M, Aaseth J, Chirumbolo S. Thymosin β4: A Multi-Faceted Tissue Repair Stimulating Protein in Heart Injury. Curr Med Chem 2021; 27:6294-6305. [PMID: 31333080 DOI: 10.2174/0929867326666190716125456] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2019] [Revised: 07/01/2019] [Accepted: 07/02/2019] [Indexed: 12/16/2022]
Abstract
Thymosin Beta-4 (Tβ4) is known as a major pleiotropic actin-sequestering protein that is involved in tumorigenesis. Tβ4 is a water-soluble protein that has different promising clinical applications in the remodeling and ulcerated tissues repair following myocardial infarction, stroke, plasticity and neurovascular remodeling of the Peripheral Nervous System (PNS) and the Central Nervous System (CNS). On the other hand, similar effects have been observed for Tβ4 in other kinds of tissues, including cardiac muscle tissue. In recent reports, as it activates resident epicardial progenitor cells and modulates inflammatory-caused injuries, Tβ4 has been suggested as a promoter of the survival of cardiomyocytes. Furthermore, Tβ4 may act in skeletal muscle and different organs in association/synergism with numerous other tissue repair stimulating factors, including melatonin and C-fiber-derived peptides. For these reasons, the present review highlights the promising role of Tβ4 in cardiac healing.
Collapse
Affiliation(s)
- Geir Bjørklund
- Council for Nutritional and Environmental Medicine, Mo i Rana, Norway
| | - Maryam Dadar
- Razi Vaccine and Serum Research Institute, Agricultural Research, Education and Extension Organization (AREEO), Karaj, Iran
| | - Jan Aaseth
- Research Department, Innlandet Hospital Trust, Brumunddal, Norway,Inland Norway University of Applied Sciences, Elverum, Norway
| | - Salvatore Chirumbolo
- Department of Neurosciences, Biomedicine and Movement Sciences,
University of Verona, Verona, Italy
| |
Collapse
|
8
|
Gezen M, Yılmaz Ü, Fazlıoğulları O, Yılmaz N, Horozoğlu C, Ergen A, Zeybek Ü. Thymosin beta-4 A/T polymorphism and acute coronary syndrome risk. ARCHIVES OF CLINICAL AND EXPERIMENTAL MEDICINE 2019. [DOI: 10.25000/acem.475094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
|
9
|
Polley A, Sen P, Sengupta A, Chakraborty S. β-Catenin stabilization promotes proliferation and increase in cardiomyocyte number in chick embryonic epicardial explant culture. In Vitro Cell Dev Biol Anim 2017; 53:922-939. [PMID: 28842809 DOI: 10.1007/s11626-017-0191-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Accepted: 07/24/2017] [Indexed: 12/28/2022]
Abstract
Cardiomyocyte (CM) differentiation from proepicardial organ- (PEO) and embryonic epicardium (eEpi)-derived cells or EPDCs in a developing heart emerges as a wide interest in purview of cardiac repair and regenerative medicine. eEpi originates from the precursor PEO and EPDCs, which contribute to several cardiac cell types including smooth muscle cells, fibroblasts, endothelial cells, and CMs during cardiogenesis. Here in this report, we have analyzed several cardiac lineage-specific marker gene expressions between PEO and eEpi cells. We have found that PEO-derived cells show increased level of CM lineage-specific marker gene expression compared to eEpi cells. Moreover, Wnt signaling activation results in increased level of CM-specific marker gene expression in both PEO and eEpi cells in culture. Interestingly, Wnt signaling activation also increases the number of proliferating and sarcomeric myosin (Mf20)-positive cells in eEpi explant culture. Together, this data suggests that eEpi cells as a source for CM differentiation and Wnt signaling mediator, β-catenin, might play an important role in CM differentiation from eEpi cells in culture.
Collapse
Affiliation(s)
- Anisha Polley
- Department of Life Sciences, Presidency University, 86/1, College Street, Baker building, 2nd floor, Kolkata, 700073, India
| | - Puja Sen
- Department of Life Sciences, Presidency University, 86/1, College Street, Baker building, 2nd floor, Kolkata, 700073, India
| | - Arunima Sengupta
- The Department of Life sciences and Biotechnology, Jadavpur University, Kolkata, 700032, India
| | - Santanu Chakraborty
- Department of Life Sciences, Presidency University, 86/1, College Street, Baker building, 2nd floor, Kolkata, 700073, India.
| |
Collapse
|
10
|
Thymosin beta-4 overexpression correlates with high-risk groups in gastric gastrointestinal stromal tumors: A retrospective analysis by immunohistochemistry. Pathol Res Pract 2017; 213:1139-1143. [PMID: 28756979 DOI: 10.1016/j.prp.2017.07.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Revised: 06/01/2017] [Accepted: 07/02/2017] [Indexed: 12/19/2022]
Abstract
BACKGROUND Thymosin beta-4 (Tβ4) is a protein that is linked to a number of important biological actions and recently tumor progression and poor prognosis of some tumors. The aim of this study was to evaluate Tβ4 expression in gastric GISTs and correlate with some clinicopathological characteristics related with prognosis and clinical outcome in order to add further data to the current literature. METHODS Tβ4 antibody was applied to the 4μm-thick paraffin sections of 57 gastric GISTs by immunohistochemistry. RESULTS Tβ4 expression was found to be directly corrrelated with higher risk groups, tumor size, mitotic count, cellularity, and necrosis while it was inversely correlated with overall survival (OS) by univariate analysis (p=0.000, p=0.001, p=0.000, p=0.025, p=0.023, and p=0.042, respectively). The direct association between Tβ4 expression and risk groups were also supported by multivariate analysis (p=0.000, β=0.497, t=4.374). CONCLUSION Overexpression of Tβ4 was found to be related with predictive characteristics for tumor progression and adverse prognosis. Thus, we suggest that overexpression of Tβ4 might play a role in the progression of gastric GISTs and might be used as a potential prognostic tool as well as a target for novel therapies.
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
|
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
|
13
|
Sofia RR, Serra AJ, Silva JA, Antonio EL, Manchini MT, Oliveira FAAD, Teixeira VPC, Tucci PJF. Gender-based differences in cardiac remodeling and ILK expression after myocardial infarction. Arq Bras Cardiol 2014; 103:124-30. [PMID: 25098374 PMCID: PMC4150663 DOI: 10.5935/abc.20140113] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2013] [Accepted: 01/17/2014] [Indexed: 01/13/2023] Open
Abstract
Background Gender can influence post-infarction cardiac remodeling. Objective To evaluate whether gender influences left ventricular (LV) remodeling and
integrin-linked kinase (ILK) after myocardial infarction (MI). Methods Female and male Wistar rats were assigned to one of three groups: sham, moderate
MI (size: 20-39% of LV area), and large MI (size: ≥40% of LV area). MI was
induced by coronary occlusion, and echocardiographic analysis was performed after
six weeks to evaluate MI size as well as LV morphology and function. Real-time
RT-PCR and Western blot were used to quantify ILK in the myocardium. Results MI size was similar between genders. MI resulted in systolic dysfunction and
enlargement of end-diastolic as well as end-systolic dimension of LV as a function
of necrotic area size in both genders. Female rats with large MI showed a lower
diastolic and systolic dilatation than the respective male rats; however, LV
dysfunction was similar between genders. Gene and protein levels of ILK were
increased in female rats with moderate and large infarctions, but only male rats
with large infarctions showed an altered ILK mRNA level. A negative linear
correlation was evident between LV dimensions and ILK expression in female rats
with large MI. Conclusions Post-MI ILK expression is altered in a gender-specific manner, and higher ILK
levels found in females may be sufficient to improve LV geometry but not LV
function.
Collapse
Affiliation(s)
- Renato Rodrigues Sofia
- Programa de Pós-graduação em Ciências da Reabilitação, Universidade Nove de Julho, São Paulo, SP, Brazil
| | - Andrey Jorge Serra
- Programa de Pós-graduação em Ciências da Reabilitação, Universidade Nove de Julho, São Paulo, SP, Brazil
| | - Jose Antonio Silva
- Programa de Pós-graduação em Ciências da Reabilitação, Universidade Nove de Julho, São Paulo, SP, Brazil
| | - Ednei Luiz Antonio
- Departamento de Cardiologia, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | - Martha Trindade Manchini
- Programa de Pós-graduação em Ciências da Reabilitação, Universidade Nove de Julho, São Paulo, SP, Brazil
| | | | | | | |
Collapse
|
14
|
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
|
15
|
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
|
16
|
Nemolato S, Cabras T, Restivo A, Zorcolo L, Di Felice E, Fanni D, Gerosa C, Messana I, Castagnola M, Faa G, Casula G. Thymosin ß4 expression in colorectal polyps and adenomas. Clinics (Sao Paulo) 2013; 68:1220-4. [PMID: 24141838 PMCID: PMC3782725 DOI: 10.6061/clinics/2013(09)08] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2013] [Accepted: 04/24/2013] [Indexed: 01/28/2023] Open
Abstract
OBJECTIVE Thymosin beta 4 (Tβ4) is a ubiquitous peptide that plays pivotal roles in the cytoskeletal system and in cell differentiation. Recently, a role for Tβ4 has been proposed in experimental and human carcinogenesis, including gastrointestinal cancer. This study was aimed at evaluating the relationship between Tβ4 immunoreactivity and the initial steps of carcinogenesis. METHODS In total, 60 intestinal biopsies, including 10 hyperplastic polyps, 10 sessile serrated adenomas/polyps, 15 colorectal adenomas with low-grade dysplasia, 15 adenomas with high-grade dysplasia, 15 adenocarcinomas and 10 samples of normal colon mucosa, were analyzed for Tβ4 expression by immunohistochemistry. RESULTS Weak cytoplasmic reactivity for Tβ4 was detected in the normal colon mucosa. No reactivity for Tβ4 was found in hyperplastic and sessile serrated polyps/adenomas. Tβ4 expression was observed in 10/15 colorectal adenocarcinomas. In adenomas with low-grade dysplasia, Tβ4 immunoreactivity was mainly detected in dysplastic glands but was absent in hyperplastic glands. Tβ4 immunoreactivity was characterized by spot-like perinuclear staining. In high-grade dysplastic polyps, immunostaining for Tβ4 appeared diffuse throughout the entire cytoplasm of dysplastic cells. Spot-like perinuclear reactivity was detected in adenocarcinoma tumor cells. CONCLUSIONS Our study shows for the first time that Tβ4 is expressed during different steps of colon carcinogenesis. The shift of Tβ4 immunolocalization from low-grade to high-grade dysplastic glands suggests a role for Tβ4 in colorectal carcinogenesis. However, the real meaning of Tβ4 reactivity in dysplastic intestinal epithelium remains unknown.
Collapse
Affiliation(s)
- Sonia Nemolato
- Department of Surgery, University of Cagliari, Cagliari, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Lv S, Cheng G, Zhou Y, Xu G. Thymosin beta4 induces angiogenesis through Notch signaling in endothelial cells. Mol Cell Biochem 2013; 381:283-90. [PMID: 23749167 DOI: 10.1007/s11010-013-1713-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2013] [Accepted: 05/28/2013] [Indexed: 10/26/2022]
Abstract
Thymosin beta4 (Tβ4) has multi-functional roles in angiogenesis and arteriogenesis, but little is known about its mechanism. The Notch signaling pathway is important in regulation of angiogenic behavior of endothelial cells, in addition to vascular endothelial growth factor (VEGF). Whether, Tβ4 regulates angiogenesis through Notch signaling pathway is not clear. In this article, we evaluated the effect of Notch signaling in Tβ4-induced angiogenesis in human umbilical vein endothelial cell (HUVEC). Our results revealed that Tβ4 increased Notch1 and Notch4 expression in a dose and time-dependent manner. The inhibition of Notch1 or Notch4 with siRNA or the Notch receptor inhibitor DAPT significantly prevented Tβ4-induced HUVEC tube formation and lymphocyte transendothelial migration. The inhibition of Notch1 or Notch4 also blocked Tβ4-induced VEGF and HIF-1α expression. VE-cadherin is the major endothelial adhesion molecule in the control of angiogenesis. Tβ4 significantly reduced VE-cadherin expression levels in HUVEC, while the inhibition of Notch signaling prevented Tβ4-induced VE-cadherin down-regulation. The results of this study suggest that Tβ4 induces HUVEC angiogenesis through Notch signaling pathway.
Collapse
Affiliation(s)
- Shumin Lv
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University College of Medicine, Hangzhou, Zhejiang, China
| | | | | | | |
Collapse
|
18
|
Tantos A, Szabo B, Lang A, Varga Z, Tsylonok M, Bokor M, Verebelyi T, Kamasa P, Tompa K, Perczel A, Buday L, Lee SH, Choo Y, Han KH, Tompa P. Multiple fuzzy interactions in the moonlighting function of thymosin-β4. INTRINSICALLY DISORDERED PROTEINS 2013; 1:e26204. [PMID: 28516021 PMCID: PMC5424802 DOI: 10.4161/idp.26204] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/29/2013] [Revised: 08/15/2013] [Accepted: 08/18/2013] [Indexed: 12/19/2022]
Abstract
Thymosine β4 (Tß4) is a 43 amino acid long intrinsically disordered protein (IDP), which was initially identified as an actin-binding and sequestering molecule. Later it was described to have multiple other functions, such as regulation of endothelial cell differentiation, blood vessel formation, wound repair, cardiac cell migration, and survival.1 The various functions of Tβ4 are mediated by interactions with distinct and structurally unrelated partners, such as PINCH, ILK, and stabilin-2, besides the originally identified G-actin. Although the cellular readout of these interactions and the formation of these complexes have been thoroughly described, no attempt was made to study these interactions in detail, and to elucidate the thermodynamic, kinetic, and structural underpinning of this range of moonlighting functions. Because Tβ4 is mostly disordered, and its 4 described partners are structurally unrelated (the CTD of stabilin-2 is actually fully disordered), it occurred to us that this system might be ideal to characterize the structural adaptability and ensuing moonlighting functions of IDPs. Unexpectedly, we found that Tβ4 engages in multiple weak, transient, and fuzzy interactions, i.e., it is capable of mediating distinct yet specific interactions without adapting stable folded structures.
Collapse
Affiliation(s)
- Agnes Tantos
- Institute of Enzymology; Research Centre for Natural Sciences; Hungarian Academy of Sciences; Budapest, Hungary
| | - Beata Szabo
- Institute of Enzymology; Research Centre for Natural Sciences; Hungarian Academy of Sciences; Budapest, Hungary
| | - Andras Lang
- Eötvös Loránd University; Institute of Chemistry; Budapest, Hungary
| | - Zoltan Varga
- Institute of Enzymology; Research Centre for Natural Sciences; Hungarian Academy of Sciences; Budapest, Hungary
| | - Maksym Tsylonok
- VIB Department of Structural Biology; Vrije Universiteit Brussel; Brussels, Belgium
| | - Monika Bokor
- Institute for Solid State Physics and Optics; Wigner Research Centre for Physics of the Hungarian Academy of Sciences; Budapest, Hungary
| | - Tamas Verebelyi
- Institute for Solid State Physics and Optics; Wigner Research Centre for Physics of the Hungarian Academy of Sciences; Budapest, Hungary
| | - Pawel Kamasa
- Institute for Solid State Physics and Optics; Wigner Research Centre for Physics of the Hungarian Academy of Sciences; Budapest, Hungary
| | - Kalman Tompa
- Institute for Solid State Physics and Optics; Wigner Research Centre for Physics of the Hungarian Academy of Sciences; Budapest, Hungary
| | - Andras Perczel
- Eötvös Loránd University; Institute of Chemistry; Budapest, Hungary
| | - Laszlo Buday
- Institute of Enzymology; Research Centre for Natural Sciences; Hungarian Academy of Sciences; Budapest, Hungary
| | - Si Hyung Lee
- Division of Biosystems Research; Korea Research Institute of Bioscience and Biotechnology; Daejeon, Republic of Korea
| | - Yejin Choo
- Division of Biosystems Research; Korea Research Institute of Bioscience and Biotechnology; Daejeon, Republic of Korea
| | - Kyou-Hoon Han
- Division of Biosystems Research; Korea Research Institute of Bioscience and Biotechnology; Daejeon, Republic of Korea
- Department of Bioinformatics; University of Science and Technology; Daejeon, Republic of Korea
| | - Peter Tompa
- Institute of Enzymology; Research Centre for Natural Sciences; Hungarian Academy of Sciences; Budapest, Hungary
- VIB Department of Structural Biology; Vrije Universiteit Brussel; Brussels, Belgium
| |
Collapse
|
19
|
Gupta S, Kumar S, Sopko N, Qin Y, Wei C, Kim IK. Thymosin β4 and cardiac protection: implication in inflammation and fibrosis. Ann N Y Acad Sci 2012; 1269:84-91. [DOI: 10.1111/j.1749-6632.2012.06752.x] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
|
20
|
Chiu LLY, Reis LA, Momen A, Radisic M. Controlled release of thymosin β4 from injected collagen-chitosan hydrogels promotes angiogenesis and prevents tissue loss after myocardial infarction. Regen Med 2012; 7:523-33. [PMID: 22817626 DOI: 10.2217/rme.12.35] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
AIMS Acute myocardial infarction (MI) leads to fibrosis and severe left ventricular wall thinning. Enhancing vascularization within the infarct reduces cell death and maintains a thick left ventricular wall, which is essential for proper cardiac function. Here, we evaluated the controlled delivery of thymosin β4 (Tβ4), which supports cardiomyocyte survival by inducing vascularization and upregulating Akt activity, in the treatment of MI. MATERIALS & METHODS We injected collagen-chitosan hydrogel with controlled release of Tβ4 into the infarct after performing left anterior descending artery ligation in rats. RESULTS Tβ4-encapsulated hydrogel (thymosin) significantly reduced tissue loss post-MI (13 ± 4%), compared with 58 ± 3% and 30 ± 8% tissue loss for no treatment (MI only) and Tβ4-free hydrogel (control). Significantly more Factor VIII-positive blood vessels with diameter >50 µm were in the thymosin group compared with both MI only and control (p < 0.0001), showing Tβ4-induced vascularization. Wall thickness was positively correlated with the mature blood vessel density (r = 0.9319; p < 0.0001). CONCLUSION Controlled release of Tβ4 within the infarct enhances angiogenesis and presence of cardiomyocytes that are necessary for cardiac repair.
Collapse
Affiliation(s)
- Loraine L Y Chiu
- Chemical Engineering & Applied Chemistry, University of Toronto, ON, Canada
| | | | | | | |
Collapse
|
21
|
Karantalis V, Balkan W, Schulman IH, Hatzistergos KE, Hare JM. Cell-based therapy for prevention and reversal of myocardial remodeling. Am J Physiol Heart Circ Physiol 2012; 303:H256-70. [PMID: 22636682 DOI: 10.1152/ajpheart.00221.2012] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Although pharmacological and interventional advances have reduced the morbidity and mortality of ischemic heart disease, there is an ongoing need for novel therapeutic strategies that prevent or reverse progressive ventricular remodeling following myocardial infarction, the process that forms the substrate for ventricular failure. The development of cell-based therapy as a strategy to repair or regenerate injured tissue offers extraordinary promise for a powerful anti-remodeling therapy. In this regard, the field of cell therapy has made major advancements in the past decade. Accumulating data from preclinical studies have provided novel insights into stem cell engraftment, differentiation, and interactions with host cellular elements, as well as the effectiveness of various methods of cell delivery and accuracy of diverse imaging modalities to assess therapeutic efficacy. These findings have in turn guided rationally designed translational clinical investigations. Collectively, there is a growing understanding of the parameters that underlie successful cell-based approaches for improving heart structure and function in ischemic and other cardiomyopathies.
Collapse
Affiliation(s)
- Vasileios Karantalis
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Florida, USA
| | | | | | | | | |
Collapse
|
22
|
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
|
23
|
Bollini S, Cheung KK, Riegler J, Dong X, Smart N, Ghionzoli M, Loukogeorgakis SP, Maghsoudlou P, Dubé KN, Riley PR, Lythgoe MF, De Coppi P. Amniotic fluid stem cells are cardioprotective following acute myocardial infarction. Stem Cells Dev 2011; 20:1985-94. [PMID: 21534857 DOI: 10.1089/scd.2010.0424] [Citation(s) in RCA: 78] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
In recent years, various types of stem cells have been characterized and their potential for cardiac regeneration has been investigated. We have previously described the isolation of broadly multipotent cells from amniotic fluid, defined as amniotic fluid stem (AFS) cells. The aim of this study was to investigate the therapeutic potential of human AFS cells (hAFS) in a model of acute myocardial infarction. Wistar rats underwent 30 min of ischemia by ligation of the left anterior descending coronary artery, followed by administration of hAFS cells and 2 h of reperfusion. Infarct size was assessed by 2,3,5-triphenyltetrazolium chloride staining and planimetry. hAFS cells were also analyzed by enzyme-linked immunosorbent assay to detect secretion of putative paracrine factors, such as the actin monomer-binding protein thymosin β4 (Tβ4). The systemic injection of hAFS cells and their conditioned medium (hAFS-CM) was cardioprotective, improving myocardial cell survival and decreasing the infarct size from 53.9%±2.3% (control animals receiving phosphate-buffered saline injection) to 40.0%±3.0% (hAFS cells) and 39.7%±2.5% (hAFS-CM, P<0.01). In addition, hAFS cells were demonstrated to secrete Tβ4, previously shown to be both cardioprotective and proangiogenic. Our results suggest that AFS cells have therapeutic potential in the setting of acute myocardial infarction, which may be mediated through paracrine effectors such as Tβ4. Therefore, AFS cells might represent a novel source for cell therapy and cell transplantation strategies in repair following ischemic heart disease, with a possible paracrine mechanism of action and a potential molecular candidate for acute cardioprotection.
Collapse
Affiliation(s)
- Sveva Bollini
- Surgery Unit, Institute of Child Health and Great Ormond Street Hospital, University College London, London, UK
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Reyes-Gordillo K, Shah R, Popratiloff A, Fu S, Hindle A, Brody F, Rojkind M. Thymosin-β4 (Tβ4) blunts PDGF-dependent phosphorylation and binding of AKT to actin in hepatic stellate cells. THE AMERICAN JOURNAL OF PATHOLOGY 2011; 178:2100-8. [PMID: 21514425 DOI: 10.1016/j.ajpath.2011.01.025] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/12/2010] [Revised: 12/22/2010] [Accepted: 01/18/2011] [Indexed: 01/18/2023]
Abstract
Hepatic stellate cell transdifferentiation is a key event in the fibrogenic cascade. Therefore, attempts to prevent and/or revert the myofibroblastic phenotype could result in novel therapeutic approaches to treat liver cirrhosis. The expression of platelet-derived growth factor (PDGF)-β receptor and the proliferative response to platelet-derived growth factor-ββ (PDGF-ββ) are hallmarks of the transdifferentiation of hepatic stellate cells (HSC). In this communication, we investigated whether thymosin-β4 (Tβ4), a chemokine expressed by HSC could prevent PDGF-BB-mediated proliferation and migration of cultured HSC. Using early passages of human HSC, we showed that Tβ4 inhibited cell proliferation and migration and prevented the expression of PDGF-β receptor (PDGF-βr), α-smooth muscle actin and α1(I) collagen mRNAs. Tβ4 also inhibited the reappearance of PDGF-βr after its PDGF-BB-dependent degradation. These PDGF-dependent events were associated with the inhibition of AKT phosphorylation at both T308 and S473 amino acid residues. The lack of AKT phosphorylation was not due to the inhibition of PDGF-βr phosphorylation, the activation of phosphoinositide 3-kinase (PI3K), pyruvate dehydrogenase kinase isozyme 1 (PDK1), and mammalian target of rapamycin (mTOR). We found that PDGF-BB induced AKT binding to actin, and that Tβ4 prevented this effect. Tβ4 also prevented the activation of freshly isolated HSC cultured in the presence of Dulbecco's modified Eagle's medium or Dulbecco's minimal essential medium containing 10% fetal bovine serum. In conclusion, overall, our findings suggest that Tβ4 by sequestering actin prevents binding of AKT, thus inhibiting its phosphorylation. Therefore, Tβ4 has the potential to be an antifibrogenic agent.
Collapse
Affiliation(s)
- Karina Reyes-Gordillo
- Department of Biochemistry and Molecular Biology, The George Washington University Medical Center, Washington, DC, USA
| | | | | | | | | | | | | |
Collapse
|
25
|
Liang X, Sun Y, Chen J. Particularly interesting cysteine- and histidine-rich protein in cardiac development and remodeling. J Investig Med 2011; 57:842-8. [PMID: 19952891 DOI: 10.2310/jim.0b013e3181c5e31d] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Integrin-mediated cell-extracellular matrix interaction plays key roles in tissue morphogenesis and integrity. The Lin11-Isl-1-Mec-3 (LIM) domain-only particularly interesting cysteine- and histidine-rich (PINCH) protein functions as an adaptor essential for the assembly and function of the focal adhesion complex that links integrin signaling to the cytoskeleton and other intracellular signaling pathways and regulates diverse cellular processes such as cell adhesion, migration, growth, differentiation, and survival. Recent biochemical and genetic studies have greatly advanced our knowledge surrounding the molecular interactions and functions of each component of the focal adhesion complex and revealed a requirement for PINCH in early embryogenesis, in morphogenesis of the neural crest and cardiac outflow, and in myocardial growth and remodeling. In this review article, we will provide an overview of the current knowledge of the molecular interactions of PINCH with other components of focal adhesions, highlighting recent discoveries of the in vivo role of PINCH and discuss its potential implication for human heart disease.
Collapse
Affiliation(s)
- Xingqun Liang
- Department of Medicine, University of California at San Diego (UCSD), La Jolla, CA 92093-0613C, USA
| | | | | |
Collapse
|
26
|
Brenner C, Franz WM. The use of stem cells for the repair of cardiac tissue in ischemic heart disease. Expert Rev Med Devices 2011; 8:209-25. [PMID: 21381911 DOI: 10.1586/erd.10.78] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Ischemic heart diseases are the leading cause of death in the Western world. With increasing numbers of patients surviving their acute myocardial infarction owing to effective heart catheter techniques and intensive care treatment, congestive heart failure has become an increasing health concern. With therapeutic options for the prevention and treatment of ischemic heart disease being limited at present, huge efforts have been made in the field of stem cell research to try to establish new approaches for myocardial tissue regeneration. Owing to their pronounced differentiation potential, pluripotent stem cells seem to represent the most promising cell source for future engineering of myocardial replacement tissue. However, several crucial hurdles regarding cell yield and purity of the cultured cardiovascular progenitor cells have still not been overcome to facilitate a clinical application today. By contrast, plenty of adult stem and progenitor cells have already been well characterized and investigated in human disease. However, all of these heterogeneous cell lines primarily seem to work in a paracrine manner on ischemic myocardial tissue, rather than transdifferentiating into contractile cardiomyocytes. This article will focus on the production, application and present limitations of stem cells potentially applicable for myocardial repair.
Collapse
Affiliation(s)
- Christoph Brenner
- Department of Internal Medicine I, Munich University Hospital, Campus Grosshadern, Marchioninistr. 15, 81377 Munich, Germany
| | | |
Collapse
|
27
|
Lee HR, Yoon SY, Song SB, Park Y, Kim TS, Kim S, Hur DY, Song HK, Park H, Cho D. Interleukin-18-mediated interferon-gamma secretion is regulated by thymosin beta 4 in human NK cells. Immunobiology 2011; 216:1155-62. [PMID: 21742406 DOI: 10.1016/j.imbio.2011.04.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2010] [Accepted: 04/06/2011] [Indexed: 12/01/2022]
Abstract
Thymosin beta 4 (Tβ4) is the major G-actin sequestering molecule and is abundant in lymphoid tissues. However, it is not clear what regulates Tβ4 expression and what its function is on natural killer (NK) cells. We investigated whether interleukin-18 (IL-18) has a role in Tβ4 expression and if enhanced Tβ4 influences IL-18-mediated interferon-gamma (IFN-γ) secretion. In this study, recombinant human IL-18 (rhIL-18) enhanced the endogenous level of Tβ4 through p38MAPK and JNK signaling pathway in the human NK cell line, NK-92MI. Overexpression of endogeneous Tβ4 stimulated IFN-γ expression and secretion. Additionally, pretreatment with an inhibitor for Tβ4 decreased IL-18-enhanced IFN-γ secretion, and transfection with Tβ4-specific short hairpin RNA resulted in reduction of IFN-γ production in primary NK cells as well as in the human NK cell line. Taken together, these data indicated that Tβ4 is regulated by IL-18 and is involved in IL-18-enhanced IFN-γ secretion in NK cells.
Collapse
Affiliation(s)
- Ha-reum Lee
- Department of Life Science, Sookmyung Women's University, Seoul, Republic of Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Shah AP, Youngquist ST, McClung CD, Tzvetkova E, Hanif MA, Rosborough JP, Niemann JT. Markers of progenitor cell recruitment and differentiation rise early during ischemia and continue during resuscitation in a porcine acute ischemia model. J Interferon Cytokine Res 2011; 31:509-13. [PMID: 21332365 DOI: 10.1089/jir.2010.0133] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Clinical administration of bone marrow-derived stem cells in the setting of acute myocardial infarction (AMI) leads to improved left ventricular ejection fraction. Thymosin beta-4 (TB4) and vascular endothelial growth factor (VEGF) are linked to adult epicardial progenitor cell mobilization and neovascularization and is cardioprotective after myocardial ischemia. This study investigated the time course of TB4 and VEGF during AMI, cardiac arrest, and resuscitation. Fifteen anesthetized and instrumented domestic swine underwent balloon occlusion of the proximal left anterior descending coronary artery. During occlusion, venous blood samples were collected from the right atrium at 5-min intervals until 15 min after the onset of cardiopulmonary resuscitation (CPR). Plasma levels of TB4, VEGF, and matrix metalloproteinase-9 (MMP-9, selected as a marker for remodeling and repair) were measured by ELISA. Generalized linear mixed models were employed to model the time-dependent change in plasma concentration. All variables were natural log transformed, except TB4 values, to normalize distributions. Fifteen animals successfully underwent balloon occlusion of the left anterior descending coronary artery and samples were collected from these subjects. The average onset of spontaneous ventricular fibrillation was 28 min. TB4, VEGF, and MMP-9 demonstrated a statistically significant, time-dependent increase in concentration during ischemia. Following arrest and throughout the first 15 min of resuscitation, MMP-9 had an unchanged rate of rise when compared with the prearrest, ischemic period, with VEGF showing a deceleration in its time-dependent concentration trajectory and TB4 demonstrating an acceleration. Endogenous TB4 and VEGF increase shortly after the onset of AMI and increase through cardiac arrest and resuscitation in parallel to remodeling proteases. These markers continue to rise during successful resuscitation and may represent an endogenous mechanism to recruit undifferentiated stem cells to areas of myocardial injury.
Collapse
Affiliation(s)
- Atman P Shah
- Division of Cardiology, Department of Medicine, University of Chicago, Chicago, IL 60637, USA.
| | | | | | | | | | | | | |
Collapse
|
29
|
Bollini S, Smart N, Riley PR. Resident cardiac progenitor cells: at the heart of regeneration. J Mol Cell Cardiol 2011; 50:296-303. [PMID: 20643135 DOI: 10.1016/j.yjmcc.2010.07.006] [Citation(s) in RCA: 96] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [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
|
30
|
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 2011; 114:102-15. [PMID: 20486893 PMCID: PMC2962722 DOI: 10.3171/2010.4.jns10118] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [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
|
31
|
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: 63] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2010] [Accepted: 09/09/2010] [Indexed: 12/12/2022]
|
32
|
Edwards J. Vertebrate beta-thymosins: conserved synteny reveals the relationship between those of bony fish and of land vertebrates. FEBS Lett 2010; 584:1047-53. [PMID: 20138884 DOI: 10.1016/j.febslet.2010.02.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2009] [Revised: 02/01/2010] [Accepted: 02/02/2010] [Indexed: 11/16/2022]
Abstract
Using conservation of synteny I show how the four thymosins expressed by teleost fish are related to the three of tetrapods, which is not evident from their protein sequences. This clarification was aided by identification of a novel thymosin of reptilians that replaces the beta10 thymosin of mammals. Recent reconstruction of the ancestral vertebrate genome suggests that divergence of beta-thymosins began with duplication preceding the two rounds of whole genome duplication.
Collapse
Affiliation(s)
- John Edwards
- Faculty of Biomedical and Life Sciences, University of Glasgow, United Kingdom.
| |
Collapse
|
33
|
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
|
34
|
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: 67] [Impact Index Per Article: 4.2] [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
|
35
|
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: 45] [Impact Index Per Article: 2.8] [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
|
36
|
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
|
37
|
Kovacic JC, Harvey RP, Dimmeler S. Cardiovascular regenerative medicine: digging in for the long haul. Cell Stem Cell 2008; 1:628-33. [PMID: 18371404 DOI: 10.1016/j.stem.2007.11.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Cardiovascular cell-based regenerative medicine has enjoyed a brief but exciting history. In little over a decade, multiple hypotheses have risen and fallen, and this work has now triggered a critical reconsideration of several long-held cardiovascular paradigms. These and other issues were the focus of the second Symposium on Cardiovascular Regenerative Medicine, recently held at the NIH-NHLBI in Bethesda, MD, USA. The meeting served to showcase some of the highlights of the past decade but, at the same time, sharply underlined the enormity of the task ahead. Collectively, a sense emerged that researchers in this field are "digging in for the long haul."
Collapse
Affiliation(s)
- Jason C Kovacic
- Cardiology Branch, Division of Intramural Research, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| | | | | |
Collapse
|