1
|
Li X, Hu H, Guo D, Hu Y, Zhou H, Chen Y, Fang X. Imbalance of Pro- and Anti-inflammatory Cytokines Induced Different Types of Recurrent Atrial Arrhythmias after Drug Eluting Coronary Stent Implantation. Curr Vasc Pharmacol 2022; 20:447-456. [PMID: 36045517 DOI: 10.2174/1570161120666220831094507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 06/08/2022] [Accepted: 07/27/2022] [Indexed: 01/25/2023]
Abstract
BACKGROUND Atrial arrhythmias are associated with an increased risk of stroke and death in the elderly. The risk and predictive factors of recurrent atrial arrhythmias in elderly patients after coronary stenting are not well known. OBJECTIVE This research sought to investigate the roles of pro- and anti-inflammatory cytokine imbalances in different types of recurrent atrial arrhythmias in elderly patients defined as individuals aged 65 years or older after sirolimus eluting stent (Cordis, Warren, New Jersey) implantation. METHODS We measured interleukin-1β (IL-1β), interleukin-6 (IL-6), interleukin-8 (IL-8), tumor necrosis factor-α (TNF-α), interleukin-10 (IL-10), interleukin-17 (IL-17), interleukin-13 (IL-13) and interleukin- 37 (IL-37) in elderly patients with recurrent atrial arrhythmias and assessed the impact of pro- and antiinflammatory cytokine imbalances on recurrent atrial arrhythmias in elderly patients after coronary stenting. RESULTS Levels of IL-1 β, IL-6, IL-8, and TNF-α were remarkably increased (p<0.001), and IL-10, IL- 17, IL-13, and IL-37 were remarkably lowered (p<0.001) in elderly patients with recurrent atrial arrhythmias after coronary stent implantation. Imbalance of pro- and anti-inflammatory cytokines induced recurrent atrial arrhythmias after coronary stenting. Pro- and anti-inflammatory cytokine imbalances may be used to identify elderly patients who have an increased risk of developing recurrent atrial arrhythmias after coronary stenting. CONCLUSION The imbalance of pro- and anti-inflammatory cytokines was associated with recurrent atrial arrhythmias in elderly patients after coronary stenting. Pro- and anti-inflammatory cytokines may be clinically useful biomarkers for predicting recurrent atrial arrhythmias in elderly patients after coronary stent implantation.
Collapse
Affiliation(s)
- Xia Li
- Xiamen Road Branch Hospital, The Affiliated Huaian Hospital of Xuzhou Medical University, Huaian 223002, China
| | - Haibo Hu
- Xiamen Road Branch Hospital, The Affiliated Huaian Hospital of Xuzhou Medical University, Huaian 223002, China
| | - Dianxuan Guo
- Department of Geriatrics, The Affiliated Huaian Hospital of Xuzhou Medical University, Huaian 223002, China
| | - Youdong Hu
- Department of Geriatrics, The Affiliated Huaian Hospital of Xuzhou Medical University, Huaian 223002, China
| | - Hualan Zhou
- Department of Geriatrics, The Affiliated Huaian Hospital of Xuzhou Medical University, Huaian 223002, China
| | - Ying Chen
- Department of Geriatrics, The Affiliated Huaian Hospital of Xuzhou Medical University, Huaian 223002, China
| | - Xiang Fang
- Department of Geriatrics, The Affiliated Huaian Hospital of Xuzhou Medical University, Huaian 223002, China
| |
Collapse
|
2
|
Rao P, Skaria R, Khalpey Z. Regenerative concepts in cardiovascular research: novel hybrid therapy for remodeling ischemic cardiomyopathy. J Thorac Dis 2018; 10:S1819-S1824. [PMID: 30034858 DOI: 10.21037/jtd.2018.01.83] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Prashant Rao
- Sarver Heart Center, Division of Cardiothoracic Surgery, University of Arizona, Tucson, AZ, USA
| | - Rinku Skaria
- Department of Surgery, Division of Cardiothoracic Surgery, University of Arizona, Tucson, AZ, USA
| | - Zain Khalpey
- Department of Surgery, Division of Cardiothoracic Surgery, University of Arizona, Tucson, AZ, USA
| |
Collapse
|
3
|
Abstract
Despite considerable advances in medicine, cardiovascular disease is still rising, with ischemic heart disease being the leading cause of death and disability worldwide. Thus extensive efforts are continuing to establish effective therapeutic modalities that would improve both quality of life and survival in this patient population. Novel therapies are being investigated not only to protect the myocardium against ischemia-reperfusion injury but also to regenerate the heart. Stem cell therapy, such as potential use of human mesenchymal stem cells and induced pluripotent stem cells and their exosomes, will make it possible not only to address molecular mechanisms of cardiac conditioning, but also to develop new therapies for ischemic heart disease. Despite the studies and progress made over the last 15 years on the use of stem cell therapy for cardiovascular disease, the efforts are still in their infancy. Even though the expectations have been high, the findings indicate that most of the clinical trials generally have been small and the results inconclusive. Because of many negative findings, there is certain pessimism that cardiac cell therapy is likely to yield any meaningful results over the next decade or so. Similar to other new technologies, early failures are not unusual and they may be followed by impressive success. Nevertheless, there has been considerable attention to safety by the clinical investigators because the adverse events of stem cell therapy have been impressively rare. In summary, although regenerative biology might not help the cardiovascular patient in the near term, it is destined to do so over the next several decades.
Collapse
Affiliation(s)
- Maia Terashvili
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI
| | - Zeljko J Bosnjak
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI.
| |
Collapse
|
4
|
Choudhery MS, Khan M, Mahmood R, Mohsin S, Akhtar S, Ali F, Khan SN, Riazuddin S. Mesenchymal stem cells conditioned with glucose depletion augments their ability to repair-infarcted myocardium. J Cell Mol Med 2012; 16:2518-2529. [PMID: 22435530 PMCID: PMC3823444 DOI: 10.1111/j.1582-4934.2012.01568.x] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2011] [Accepted: 02/28/2012] [Indexed: 02/05/2023] Open
Abstract
Mesenchymal stem cells (MSCs) are an attractive candidate for autologous cell therapy, but their ability to repair damaged myocardium is severely compromised with advanced age. Development of viable autologous cell therapy for treatment of heart failure in the elderly requires the need to address MSC ageing. In this study, MSCs from young (2 months) and aged (24 months) C57BL/6 mice were characterized for gene expression of IGF-1, FGF-2, VEGF, SIRT-1, AKT, p16(INK4a) , p21 and p53 along with measurements of population doubling (PD), superoxide dismutase (SOD) activity and apoptosis. Aged MSCs displayed senescent features compared with cells isolated from young animals and therefore were pre-conditioned with glucose depletion to enhance age affected function. Pre-conditioning of aged MSCs led to an increase in expression of IGF-1, AKT and SIRT-1 concomitant with enhanced viability, proliferation and delayed senescence. To determine the myocardial repair capability of pre-conditioned aged MSCs, myocardial infarction (MI) was induced in 24 months old C57BL/6 wild type mice and GFP expressing untreated and pre-conditioned aged MSCs were transplanted. Hearts transplanted with pre-conditioned aged MSCs showed increased expression of paracrine factors, such as IGF-1, FGF-2, VEGF and SDF-1α. This was associated with significantly improved cardiac performance as measured by dp/dt(max), dp/dt(min), LVEDP and LVDP, declined left ventricle (LV) fibrosis and apoptosis as measured by Masson's Trichrome and TUNEL assays, respectively, after 30 days of transplantation. In conclusion, pre-conditioning of aged MSCs with glucose depletion can enhance proliferation, delay senescence and restore the ability of aged cells to repair senescent infarcted myocardium.
Collapse
Affiliation(s)
| | | | - Ruhma Mahmood
- National Center of Excellence in Molecular Biology, University of the PunjabLahore, Pakistan
| | - Sadia Mohsin
- National Center of Excellence in Molecular Biology, University of the PunjabLahore, Pakistan
| | - Shoaib Akhtar
- National Center of Excellence in Molecular Biology, University of the PunjabLahore, Pakistan
| | - Fatima Ali
- National Center of Excellence in Molecular Biology, University of the PunjabLahore, Pakistan
| | - Shaheen N Khan
- National Center of Excellence in Molecular Biology, University of the PunjabLahore, Pakistan
| | - Sheikh Riazuddin
- National Center of Excellence in Molecular Biology, University of the PunjabLahore, Pakistan
| |
Collapse
|
5
|
Hoke NN, Salloum FN, Kass DA, Das A, Kukreja RC. Preconditioning by phosphodiesterase-5 inhibition improves therapeutic efficacy of adipose-derived stem cells following myocardial infarction in mice. Stem Cells 2012; 30:326-35. [PMID: 22102597 DOI: 10.1002/stem.789] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The rationale of this article is enhancing the therapeutic potential of stem cells in ischemic microenvironments by novel preconditioning strategies is critical for improving cellular therapy. We tested the hypothesis that inhibition of phosphodiesterase-5 (PDE-5) with sildenafil (Viagra) or knockdown with a silencing vector in adipose-derived stem cells (ASCs) would improve their survival and enhance cardiac function following myocardial implantation in vivo. ASCs were treated with sildenafil or PDE-5 silencing vector short hairpin RNA (shRNA(PDE-5)) and subjected to simulated ischemia/reoxygenation in vitro. Both sildenafil and shRNA(PDE-5) significantly improved viability, decreased necrosis, apoptosis, and enhanced the release of growth factors, vascular endothelial growth factor (VEGF), basic fibroblast growth factor (b-FGF), and insulin-like growth factor. Inhibition of protein kinase G reversed these effects. To show the beneficial effect of preconditioned ASCs in vivo, adult male CD-1 mice underwent myocardial infarction. Preconditioned ASCs (4 × 10(5)) were directly injected intramyocardially. Preconditioned ASC-treated hearts showed consistently superior cardiac function when compared with nonpreconditioned ASCs after 4 weeks of treatment. This was associated with significantly reduced fibrosis, increased vascular density, and decreased resident myocyte apoptosis when compared with mice receiving nonpreconditioned ASCs. VEGF, b-FGF, and Angiopoietin-1 were also significantly elevated 4 weeks after cell therapy with preconditioned ASCs. We conclude that preconditioning by inhibition of PDE-5 can be a powerful novel approach to improve stem cell therapy following myocardial infarction.
Collapse
Affiliation(s)
- Nicholas N Hoke
- Pauley Heart Center, Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, Richmond, Virginia 23298, USA
| | | | | | | | | |
Collapse
|
6
|
Excitation-contraction coupling in ventricular myocytes is enhanced by paracrine signaling from mesenchymal stem cells. J Mol Cell Cardiol 2012; 52:1249-56. [PMID: 22465692 DOI: 10.1016/j.yjmcc.2012.03.008] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2011] [Revised: 02/14/2012] [Accepted: 03/12/2012] [Indexed: 12/17/2022]
Abstract
In clinical trials mesenchymal stem cells (MSCs) are transplanted into cardiac ischemic regions to decrease infarct size and improve contractility. However, the mechanism and time course of MSC-mediated cardioprotection are incompletely understood. We tested the hypothesis that paracrine signaling by MSCs promotes changes in cardiac excitation-contraction (EC) coupling that protects myocytes from cell death and enhances contractility. Isolated mouse ventricular myocytes (VMs) were treated with control tyrode, MSC conditioned-tyrode (ConT) or co-cultured with MSCs. The Ca handling properties of VMs were monitored by laser scanning confocal microscopy and whole cell voltage clamp. ConT superfusion of VMs resulted in a time dependent increase of the Ca transient amplitude (ConT(15min): ΔF/F(0)=3.52±0.38, n=14; Ctrl(15min): ΔF/F(0)=2.41±0.35, n=14) and acceleration of the Ca transient decay (τ: ConT: 269±18ms n=14; vs. Ctrl: 315±57ms, n=14). Voltage clamp recordings confirmed a ConT induced increase in I(Ca,L) (ConT: -5.9±0.5 pA/pF n=11; vs. Ctrl: -4.04±0.3 pA/pF, n=12). The change of τ resulted from increased SERCA activity. Changes in the Ca transient amplitude and τ were prevented by the PI3K inhibitors Wortmannin (100nmol/L) and LY294002 (10μmol/L) and the Akt inhibitor V (20μmol/L) indicating regulation through PI3K signal transduction and Akt activation which was confirmed by western blotting. A change in τ was also prevented in eNOS(-/-) myocytes or by inhibition of eNOS suggesting an NO mediated regulation of SERCA activity. Since paracrine signaling further resulted in increased survival of VMs we propose that the Akt induced change in Ca signaling is also a mechanism by which MSCs mediate an anti-apoptotic effect.
Collapse
|
7
|
Rose K, Gast RE, Seeger A, Krieglstein J, Klumpp S. ATP-dependent stabilization and protection of fibroblast growth factor 2. J Biotechnol 2010; 145:54-9. [PMID: 19836424 DOI: 10.1016/j.jbiotec.2009.10.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2009] [Revised: 10/02/2009] [Accepted: 10/08/2009] [Indexed: 12/26/2022]
Abstract
Fibroblast growth factor 2 (FGF2) plays a pivotal role in cell proliferation, angiogenesis and neuroprotection. Several clinical trials using this growth factor in bone regeneration, wound healing and cardioprotection are initiated but the inadequate stability of FGF2 after application is one major problem. Binding of ATP to FGF2 and other growth factors has been demonstrated recently. Here we report that ATP, other nucleoside triphosphates and sodium triphosphate protect FGF2 from trypsin, plasmin and neutrophile elastase digestion in vitro. A molar ratio of 2:1 (ligand/FGF2) is sufficient for these protective effects. ADP shows only little, AMP no stabilizing effect on FGF2 indicating that the number of phosphate residues is important. Protection of FGF2 by ATP can be abolished by the addition of alkaline phosphatase hydrolyzing free and FGF2-bound ATP. The mutant FGF2 (K128A/R129A/K134A/K144A) with strongly reduced ATP-binding capacity revealed no detectable protease resistance after incubation with ATP. Furthermore, a stabilizing effect of ATP on FGF2 could also be demonstrated in cell culture experiments. ATP bound to FGF2 increased FGF2-dependent human umbilical vein endothelial cells proliferation when the growth factor was treated with neutrophile elastase or heat. For the first time these data demonstrate protection of FGF2 by bound ATP, other nucleoside triphosphates or sodium triphosphate from rapid protease digestion. Our data provide new evidence that nucleoside triphosphates are capable of protecting FGF2 and favours such stabilization for various, especially medical applications.
Collapse
Affiliation(s)
- Karsten Rose
- Institut für Pharmazeutische und Medizinische Chemie, Westfälische Wilhelms-Universität Münster, Germany.
| | | | | | | | | |
Collapse
|
8
|
Xiao J, Lv Y, Lin S, Jin L, Zhang Y, Wang X, Ma J, Hu K, Feng W, Cai L, Li X, Tan Y. Cardiac protection by basic fibroblast growth factor from ischemia/reperfusion-induced injury in diabetic rats. Biol Pharm Bull 2010; 33:444-449. [PMID: 20190407 DOI: 10.1248/bpb.33.444] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2024]
Abstract
Diabetes impairs the expression and function of endogenous growth factors, leading to increased cardiovascular events in diabetic patients. Supplementation of fibroblast growth factors (FGFs) protected the heart from ischemia/reperfusion (I/R)-induced injury in animal models. However, it has not yet been tested in diabetic heart. The present study was thus to clarify whether basic fibroblast growth factor (bFGF) could protect the heart from I/R-induced damage under diabetic conditions using a rat model. Male Sprague Dawley rats were used to induce diabetes by intraperitoneal injection of streptozotocin. Eight weeks later, I/R injury was generated in diabetic rats and age-matched non-diabetic rats. All I/R rats were administrated bFGF or saline through intramyocardial injection. Seven days after I/R, cardiac infarction, structural changes, cell death and blood vessel density, serum malondialdehyde (MDA) and cardiac enzyme lactate dehydrogenase (LDH) were examined. We found that I/R induced significant increases in the cardiac infarction, blood MDA contents and LDH activities, and the expression of caspase-3. Treatment of I/R rats with bFGF simultaneously with reperfusion significantly attenuated I/R-induced pathological changes, along with a significant increase in the cardiac blood vessel density in both diabetic and non-diabetic rates. The protective effects of bFGF on I/R-induced cardiac injury in diabetic group are less than those in non-diabetic group. The results indicated that bFGF provide a protection of the heart against I/R-induced oxidative damage, cell death and infarction under diabetic conditions.
Collapse
Affiliation(s)
- Jian Xiao
- Chinese-American Research Institute for Diabetic Complications, Wenzhou Medical College, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
9
|
Hoke N, Salloum F, Loesser-Casey K, Kukreja R. Cardiac regenerative potential of adipose tissue-derived stem cells. ACTA ACUST UNITED AC 2009; 96:251-65. [DOI: 10.1556/aphysiol.96.2009.3.1] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
10
|
Unur E, Ulger H, Ekinci N, Hacıaliogulları M, Ertekin T, Kilic E. Effect of Anti-Basic Fibroblast Growth Factor (Anti-bFGF) on In VitroEmbryonic Development in Rat. Anat Histol Embryol 2009; 38:241-5. [DOI: 10.1111/j.1439-0264.2009.00927.x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
11
|
Role of paracrine factors in stem and progenitor cell mediated cardiac repair and tissue fibrosis. FIBROGENESIS & TISSUE REPAIR 2008; 1:4. [PMID: 19014650 PMCID: PMC2584012 DOI: 10.1186/1755-1536-1-4] [Citation(s) in RCA: 138] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/20/2008] [Accepted: 10/13/2008] [Indexed: 12/14/2022]
Abstract
A new era has begun in the treatment of ischemic disease and heart failure. With the discovery that stem cells from diverse organs and tissues, including bone marrow, adipose tissue, umbilical cord blood, and vessel wall, have the potential to improve cardiac function beyond that of conventional pharmacological therapy comes a new field of research aiming at understanding the precise mechanisms of stem cell-mediated cardiac repair. Not only will it be important to determine the most efficacious cell population for cardiac repair, but also whether overlapping, common mechanisms exist. Increasing evidence suggests that one mechanism of action by which cells provide tissue protection and repair may involve paracrine factors, including cytokines and growth factors, released from transplanted stem cells into the surrounding tissue. These paracrine factors have the potential to directly modify the healing process in the heart, including neovascularization, cardiac myocyte apoptosis, inflammation, fibrosis, contractility, bioenergetics, and endogenous repair.
Collapse
|
12
|
Shi RZ, Li QP. Improving outcome of transplanted mesenchymal stem cells for ischemic heart disease. Biochem Biophys Res Commun 2008; 376:247-50. [PMID: 18789897 DOI: 10.1016/j.bbrc.2008.09.004] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2008] [Accepted: 09/02/2008] [Indexed: 01/09/2023]
Abstract
Mesenchymal stem cells (MSCs) transplantation has been proposed as a promising means for the repair and regeneration of heart cells in ischemia heart disease. However, advancement in stem cell therapy is hindered by the poor survival of implanted cells. Recently, many kinds of methods have been developed to prevent stem cells apoptosis and improve their therapeutic potential in the ischemia heart, including genetically modifying, retaining viability in vitro, suitable transplantation means, and preconditioning. Despite the great promise of these methods, there are still many problems to be solved in this field, including limited kinds of experimental animals, few clinical trials, and cell differentiation.
Collapse
Affiliation(s)
- Rui-Zhen Shi
- Department of Pharmacology, Nanjing Medical University, 140 Hanzhong Road, Nanjing 210029, China
| | | |
Collapse
|
13
|
Hu Z, Zhang F, Yang Z, Yang N, Zhang D, Zhang J, Cao K. Combination of simvastatin administration and EPC transplantation enhances angiogenesis and protects against apoptosis for hindlimb ischemia. J Biomed Sci 2008; 15:509-17. [DOI: 10.1007/s11373-008-9243-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2007] [Accepted: 02/22/2008] [Indexed: 02/02/2023] Open
|
14
|
Vogt A, Lutz S, Rümenapp U, Han L, Jakobs KH, Schmidt M, Wieland T. Regulator of G-protein signalling 3 redirects prototypical Gi-coupled receptors from Rac1 to RhoA activation. Cell Signal 2007; 19:1229-37. [PMID: 17300916 DOI: 10.1016/j.cellsig.2007.01.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2006] [Revised: 01/08/2007] [Accepted: 01/08/2007] [Indexed: 11/19/2022]
Abstract
The small GTPases, Rac1 and RhoA, are pivotal regulators of several essential, but distinct cellular processes. Numerous G-protein-coupled receptors signal to these GTPases, but with different specificities. Specifically, Gi-coupled receptors (GiPCRs) are generally believed to activate Rac1, but not RhoA, a process involving Gbetagamma-dimers and phosphatidylinositol 3-kinase (PI3K). Here we show that, depending on the expression level of the 519 amino acid isoform of regulator of G-protein signalling 3 (RGS3L), prototypical GiPCRs, like M2 muscarinic, A1 adenosine, and alpha2-adrenergic receptors, activate either Rac1 or RhoA in human embryonic kidney cells and neonatal rat cardiomyocyte-derived H10 cells. The switch from Rac1 to RhoA activation in H10 cells was controlled by fibroblast growth factor-2 (FGF-2), lowering the expression of RGS3L. Activation of both, Rac1 and RhoA, seen at low and high expression levels of RGS3L, respectively, was sensitive to pertussis toxin and the PI3K inhibitor LY294002 and mediated by Gbetagamma-dimers. We conclude that RGS3L functions as a molecular switch, redirecting GiPCRs via Gbetagamma-dimers and PI3K from Rac1 to RhoA activation. Considering the essential roles of Rac1 and RhoA in many signalling pathways, this additional function of RGS3L indicates a specific role of this protein in cellular signalling networks.
Collapse
Affiliation(s)
- Andreas Vogt
- Department of Experimental and Clinical Pharmacology and Toxicology, University of Heidelberg, Mannheim, Germany
| | | | | | | | | | | | | |
Collapse
|
15
|
Kardami E, Detillieux K, Ma X, Jiang Z, Santiago JJ, Jimenez SK, Cattini PA. Fibroblast growth factor-2 and cardioprotection. Heart Fail Rev 2007; 12:267-77. [PMID: 17516168 DOI: 10.1007/s10741-007-9027-0] [Citation(s) in RCA: 89] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Boosting myocardial resistance to acute as well as chronic ischemic damage would ameliorate the detrimental effects of numerous cardiac pathologies and reduce the probability of transition to heart failure. Experimental cardiology has pointed to ischemic and pharmacological pre- as well as post-conditioning as potent acute cardioprotective manipulations. Additional exciting experimental strategies include the induction of true regenerative and/or angiogenic responses to the damaged heart, resulting in sustained structural and functional beneficial effects. Fibroblast growth factor-2 (FGF-2), an endogenous multifunctional protein with strong affinity for the extracellular matrix and basal lamina and well-documented paracrine, autocrine and intracellular modes of action, has been shown over the years to exert acute and direct pro-survival effects, irrespectively of whether it is administered before, during or after an ischemic insult to the heart. FGF-2 is also a potent angiogenic protein and a crucial agent for the proliferation, expansion, and survival of several cell types including those with stem cell properties. Human clinical trials have pointed to a good safety record for this protein. In this review, we will present a case for the low molecular weight isoform of fibroblast growth factor-2 (lo-FGF-2) as a very promising therapeutic agent to achieve powerful acute as well as sustained benefits for the heart, due to its cytoprotective and regenerative properties.
Collapse
Affiliation(s)
- Elissavet Kardami
- Institute of Cardiovascular Sciences, St. Boniface Research Centre, University of Manitoba, 351 Taché Avenue, Winnipeg, Manitoba, Canada.
| | | | | | | | | | | | | |
Collapse
|
16
|
Xu M, Uemura R, Dai Y, Wang Y, Pasha Z, Ashraf M. In vitro and in vivo effects of bone marrow stem cells on cardiac structure and function. J Mol Cell Cardiol 2006; 42:441-8. [PMID: 17187821 PMCID: PMC1899533 DOI: 10.1016/j.yjmcc.2006.10.009] [Citation(s) in RCA: 150] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2006] [Revised: 09/15/2006] [Accepted: 10/12/2006] [Indexed: 01/02/2023]
Abstract
It is hypothesized that the protection of bone marrow stem cells (BMSCs) on ischemic myocardium might be related to the anti-apoptotic effect via paracrine mechanisms. In this study, a wide array of cytokines including vascular endothelial growth factor (VEGF), basic fibroblast growth factor (bFGF), stromal cell-derived factor-1 (SDF-1) and insulin growth factor-1 (IGF-1) were detected in the BMSCs cultured medium by ELISA. Myocyte apoptosis was assayed by DNA fragmentation and annexin-V staining. Myocardial infarction model was produced by ligation of mouse left anterior descending coronary artery (LAD). Before LAD ligation, mice were myoablated by irradiation and transplanted with bone marrow cells from transgenic mice expressing green fluorescent protein (GFP). After LAD ligation, animals were administered stem cell factor (SCF, 200 mug/day/kg, i.p.) or saline for 6 days. Animals were sacrificed at 4 weeks after SCF treatment. Apoptotic cardiomyocytes were analyzed by TUNEL. Myocardial function was analyzed by echocardiography and pressure-volume system. Bcl-2 protein was analyzed by Western blotting. Our results showed that cultured BMSCs released VEGF, bFGF, SDF-1 and IGF-1. Hypoxia-induced cell apoptosis was diminished in cardiomyocytes co-cultured with BMSCs. Smaller LV dimension and increased LV ejection fraction were seen in SCF-treated animals. SCF significantly reduced cardiomyocytes apoptosis within peri-infarct area and increased up-regulation expression of Bcl-2 in ischemic area. Moreover, conditioned medium from cultured BMSCs also induced up-regulation of Bcl-2 protein in cardiomyocytes. It is concluded that paracrine mediators secreted by BMSCs might be involved in early repair of ischemic heart by preventing cardiomyocytes apoptosis and improving cardiac function.
Collapse
Affiliation(s)
- Meifeng Xu
- Department of Pathology and Laboratory Medicine, University of Cincinnati Medical Center, 231 Albert Sabin Way, Cincinnati, OH 45267, USA
| | | | | | | | | | | |
Collapse
|
17
|
Jiang ZS, Jeyaraman M, Wen GB, Fandrich RR, Dixon IMC, Cattini PA, Kardami E. High- but not low-molecular weight FGF-2 causes cardiac hypertrophy in vivo; possible involvement of cardiotrophin-1. J Mol Cell Cardiol 2006; 42:222-33. [PMID: 17045289 DOI: 10.1016/j.yjmcc.2006.09.002] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2006] [Revised: 08/21/2006] [Accepted: 09/01/2006] [Indexed: 12/22/2022]
Abstract
The heart expresses high and low molecular weight (hmw, lmw) fibroblast growth factor 2 (FGF-2) isoforms. While the injury-repair-related activities of lmw-FGF-2 have been studied extensively, those of hmw-FGF-2 have not. Thus, we investigated the effects of hmw-FGF-2 on acute as well as chronic responses to myocardial infarction (MI) induced by irreversible coronary occlusion in the rat. Hmw- or lmw-FGF-2 was injected into the ischemic zone during acute evolving MI. Both isoforms were equally effective in reducing infarct size (at 24 h post-MI) and improving heart function up to 6 weeks post-MI, compared to a vehicle-treated infarcted group. Lmw-FGF-2 alone upregulated vascularization in the infarct. Hmw-FGF-2 elicited significant hypertrophy, compared to the vehicle-treated group, at 4-8 weeks post-MI, assessed by ultrasound, heart morphometry and cardiomyocyte cross-sectional area. In addition, hmw- (but not lmw-) FGF-2-treated hearts displayed increased accumulation of the cytokine cardiotrophin-1 and its signal transducer gp130. In culture, hmw- (but not lmw-) FGF-2 increased cardiomyocyte protein synthesis and cell size as well as upregulated cardiotrophin-1 released by cardiac fibroblasts, pointing to similar activities in vivo. Thus, hmw- and lmw-FGF-2 exert isoform-specific effects in the heart and only hmw-FGF-2 triggers cardiomyocyte hypertrophic growth. Direct effects of hmw-FGF-2 on cardiomyocytes, becoming reinforced and sustained by upregulation of cardiotrophin-1 and acting in concert with other factors, are likely to contribute to post-MI hypertrophy.
Collapse
Affiliation(s)
- Zhi-Sheng Jiang
- Institute of Cardiovascular Disease, Nanhua University, Hengyang City, Hunan, PR China
| | | | | | | | | | | | | |
Collapse
|
18
|
Villanueva S, Cespedes C, Gonzalez A, Vio CP. bFGF induces an earlier expression of nephrogenic proteins after ischemic acute renal failure. Am J Physiol Regul Integr Comp Physiol 2006; 291:R1677-87. [PMID: 16873559 DOI: 10.1152/ajpregu.00023.2006] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Recovery from acute renal failure (ARF) requires the replacement of injured cells with new cells that restore tubule epithelial integrity. We described recently the expression of a wide range of nephrogenic proteins in tubular cells after ARF induced by ischemia-reperfusion (I/R) (Villanueva S, Cespedes C, and Vio CP. Am J Physiol Regul Integr Comp Physiol 290: R861-R870, 2006). These markers, namely, Vimentin, neural cell adhesion molecules (Ncam), basic fibroblast growth factor (bFGF), paired homeobox-2 (Pax-2), bone morphogene protein-7 (BMP-7), Noggin, Lim-1, Engrailed, Smad, phospho-Smad, hypoxia-induced factor-1alpha (HIF-1alpha), VEGF, and Tie-2, are expressed in a time frame similar to that observed in normal kidney development. bFGF participates in early kidney development as a morphogen involved in mesenchyme/epithelial transition, and it is reexpressed in the recovery phase of ARF. To test the hypothesis that bFGF can accelerate the regeneration after renal damage, we used recombinant bFGF and studied the expression pattern of the above described morphogens in ARF. Male Sprague-Dawley rats were subjected to 30 min of renal ischemic injury and were injected with bFGF 30 microg/kg followed by reperfusion. Rats were killed and the expression of nephrogenic proteins were analyzed by immunohistochemistry and Western blot analysis. In the animals subjected to I/R treated with bFGF, we observed a 12- to 24-h earlier and more abundant reexpression of the proteins Ncam, bFGF, Pax-2, BMP-7, Noggin, Lim-1, Engrailed, VEGF, and Tie-2 than the I/R untreated rats. In addition, we observed a reduction in renal damage markers ED-1 and alpha-smooth muscle actin. These results indicate that bFGF can participate in the regeneration process and suggest that the treatment with bFGF can induce an earlier regeneration process after ischemic acute renal failure.
Collapse
Affiliation(s)
- Sandra Villanueva
- Dept. de Fisiologia, Pontificia Universidad Catolica de Chile, Casilla 114-D, Santiago, Chile.
| | | | | | | |
Collapse
|
19
|
Fukuhara S, Tomita S, Nakatani T, Fujisato T, Ohtsu Y, Ishida M, Yutani C, Kitamura S. Bone marrow cell-seeded biodegradable polymeric scaffold enhances angiogenesis and improves function of the infarcted heart. Circ J 2005; 69:850-7. [PMID: 15988112 DOI: 10.1253/circj.69.850] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
BACKGROUND The present study examined whether a bioengineered polyglycolic acid cloth (PGAC) impregnated with bone marrow cells (BMC) improved the function and angiogenesis of the infarcted heart. METHODS AND RESULTS The coronary artery was ligated in Lewis rats and the infarcted area was covered with a PGAC in group 1 (n=8), with a PGAC containing basic-fibroblast growth factor (b-FGF) in group 2 (n=11) and a PGAC containing b-FGF and freshly isolated BMC in group 3 (n=10). In addition, BMC derived from transgenic mice expressing green fluorescent protein (GFP)-BMC were seeded into a PGAC, which was sutured over the infarcted area of C57BL/6 mice (n=5). In the rat study, developed and systolic pressures, dp/dt max and dp/dt min) were the highest in group 3, as were the capillary density in the PGAC and infarcted area. In the mouse study, there were few GFP-BMC in the PGAC, but none in the infarcted area. CONCLUSIONS A PGAC with BMC improved cardiac function by inducing angiogenesis without migration of BMC. Freshly isolated BMC work as angiogenic inducers and a PGAC is useful as a "drug delivery system".
Collapse
Affiliation(s)
- Shinya Fukuhara
- Department of Regenerative Medicin, National Cardiovascular Center, Suita, Japan
| | | | | | | | | | | | | | | |
Collapse
|