Skip to main content
Top
Published in: American Journal of Cardiovascular Drugs 5/2006

01-09-2006 | Current Opinion

Therapeutic Potential of Thymosin-β4 and its Derivative N-acetyl-seryl-aspartyl-lysyl-proline (Ac-SDKP) in Cardiac Healing After Infarction

Author: Dr Maria A. Cavasin

Published in: American Journal of Cardiovascular Drugs | Issue 5/2006

Login to get access

Abstract

Despite the numerous advances made in the prevention and treatment of cardiovascular diseases, there is a need for new strategies to repair and/or regenerate the myocardium after ischemia and infarction in order to prevent maladaptive remodeling and cardiac dysfunction. This article compiles and analyzes the available experimental data regarding the potential therapeutic effects of thymosin-β4 and its derivative N-acetyl-seryl-aspartyl-lysyl-proline (Ac-SDKP) in cardiac healing after myocardial infarction (MI) as well as discussing the possible mechanisms involved. The healing properties of thymosin-β4 have been described in different types of tissues, such as the skin and cornea, and more recently it has been shown that thymosin-β4 facilitates cardiac repair after infarction by promoting cell migration and myocyte survival. Additionally, the tetrapeptide Ac-SDKP was reported to reduce left ventricular fibrosis in hypertensive rats, reverse fibrosis and inflammation in rats with MI, and stimulate both in vitro and in vivo angiogenesis. Ac-SDKP also reduced cardiac rupture rate in mice post-MI. Some of the effects of Ac-SDKP, such as the enhancement of angiogenesis and the decrease in inflammation and collagenase activity, are similar to those described for thymosin-β4. Thus, it is possible that Ac-SDKP could be mediating some of the beneficial effects of its precursor. Although the experimental evidence is very promising, there are no data available from a clinical trial supporting the use of thymosin-β4 or Ac-SDKP as means of healing the myocardium after MI in patients.
Literature
2.
go back to reference Hochman JS, Bulkley BH. Expansion of acute myocardial infarction: an experimental study. Circulation 1982; 65: 1446–50PubMedCrossRef Hochman JS, Bulkley BH. Expansion of acute myocardial infarction: an experimental study. Circulation 1982; 65: 1446–50PubMedCrossRef
3.
go back to reference Schuster EH, Bulkley BH. Expansion of transmural myocardial infarction: a pathophysiologic factor in cardiac rupture. Circulation 1979; 60: 1532–8PubMedCrossRef Schuster EH, Bulkley BH. Expansion of transmural myocardial infarction: a pathophysiologic factor in cardiac rupture. Circulation 1979; 60: 1532–8PubMedCrossRef
4.
go back to reference Wehrens XHT, Doevendans PA. Cardiac rupture complicating myocardial infarction. Int J Cardiol 2004; 95: 285–92PubMedCrossRef Wehrens XHT, Doevendans PA. Cardiac rupture complicating myocardial infarction. Int J Cardiol 2004; 95: 285–92PubMedCrossRef
5.
go back to reference Antman EM, Braunwald E. Acute myocardial infarction. In: Braunwald E, Fauci AS, Kasper DL, et al., editors. Harrison’s principles of internal medicine. New York: McGraw-Hill, 2001: 1386–99 Antman EM, Braunwald E. Acute myocardial infarction. In: Braunwald E, Fauci AS, Kasper DL, et al., editors. Harrison’s principles of internal medicine. New York: McGraw-Hill, 2001: 1386–99
6.
go back to reference Figueras J, Cortadellas J, Soler-Soler J. Left ventricular free wall rupture: clinical presentation and management. Heart 2000; 83: 499–504PubMedCrossRef Figueras J, Cortadellas J, Soler-Soler J. Left ventricular free wall rupture: clinical presentation and management. Heart 2000; 83: 499–504PubMedCrossRef
7.
go back to reference Bock-Marquette I, Saxena A, White MD, et al. Thymosin β4 activates integrinlinked kinase and promotes cardiac cell migration, survival and cardiac repair. Nature 2004; 432: 466–72PubMedCrossRef Bock-Marquette I, Saxena A, White MD, et al. Thymosin β4 activates integrinlinked kinase and promotes cardiac cell migration, survival and cardiac repair. Nature 2004; 432: 466–72PubMedCrossRef
8.
go back to reference Low TLK, Goldstein AL. Chemical characterization of thymosin β4. J Biol Chem 1982; 257: 1000–6PubMed Low TLK, Goldstein AL. Chemical characterization of thymosin β4. J Biol Chem 1982; 257: 1000–6PubMed
9.
go back to reference Safer D, Golla R, Nachmias VT. Isolation of a 5-kilodalton actin-sequestering peptide from human blood platelets. Proc Natl Acad Sci U S A 1990; 87: 2536–40PubMedCrossRef Safer D, Golla R, Nachmias VT. Isolation of a 5-kilodalton actin-sequestering peptide from human blood platelets. Proc Natl Acad Sci U S A 1990; 87: 2536–40PubMedCrossRef
10.
go back to reference Malinda KM, Sidhu GS, Mani H, et al. Thymosin β4 accelerates wound healing. J Invest Dermatol 1999; 113: 364–8PubMedCrossRef Malinda KM, Sidhu GS, Mani H, et al. Thymosin β4 accelerates wound healing. J Invest Dermatol 1999; 113: 364–8PubMedCrossRef
11.
go back to reference Philp D, Badamchian M, Scheremeta B, et al. Thymosin β4 and a synthetic peptide containing its actin-binding domain promote dermal wound repair in db/db diabetic mice and in aged mice. Wound Repair Regen 2003; 11: 19–24PubMedCrossRef Philp D, Badamchian M, Scheremeta B, et al. Thymosin β4 and a synthetic peptide containing its actin-binding domain promote dermal wound repair in db/db diabetic mice and in aged mice. Wound Repair Regen 2003; 11: 19–24PubMedCrossRef
12.
go back to reference Sosne G, Szliter EA, Barrett R, et al. Thymosin beta 4 promotes corneal wound healing and decreases inflammation in vivo following alkali injury. Exp Eye Res 2002; 74: 293–9PubMedCrossRef Sosne G, Szliter EA, Barrett R, et al. Thymosin beta 4 promotes corneal wound healing and decreases inflammation in vivo following alkali injury. Exp Eye Res 2002; 74: 293–9PubMedCrossRef
13.
go back to reference Gomez-Marquez J, Franco del Amo F, Carpintero P, et al. High levels of mouse thymosin beta4 mRNA in differentiating P19 embryonic cells and during development of cardiovascular tissues. Biochim Biophys Acta 1996; 1306: 187–93PubMedCrossRef Gomez-Marquez J, Franco del Amo F, Carpintero P, et al. High levels of mouse thymosin beta4 mRNA in differentiating P19 embryonic cells and during development of cardiovascular tissues. Biochim Biophys Acta 1996; 1306: 187–93PubMedCrossRef
14.
go back to reference Van den Hoff MJ, Moorman AF, Ruijter JM, et al. Myocardialization of the cardiac outflow tract. Dev Biol 1999; 212: 477–90PubMedCrossRef Van den Hoff MJ, Moorman AF, Ruijter JM, et al. Myocardialization of the cardiac outflow tract. Dev Biol 1999; 212: 477–90PubMedCrossRef
15.
go back to reference Datta SR, Dudek H, Tao X, et al. Akt phosphorylation of BAD couples survival signals to the cell-intrinsic death machinery. Cell 1997; 91: 231–41PubMedCrossRef Datta SR, Dudek H, Tao X, et al. Akt phosphorylation of BAD couples survival signals to the cell-intrinsic death machinery. Cell 1997; 91: 231–41PubMedCrossRef
16.
go back to reference Cardone MH, Roy N, Stennicke HR, et al. Regulation of cell death protease caspase-9 by phosphorylation. Science 1998; 287: 1318–21CrossRef Cardone MH, Roy N, Stennicke HR, et al. Regulation of cell death protease caspase-9 by phosphorylation. Science 1998; 287: 1318–21CrossRef
17.
go back to reference Brunet A, Bonni A, Zigmond MJ, et al. Akt promotes cell survival by phosphorylating and inhibiting a forkhead transcription factor. Cell 1999; 96: 857–68PubMedCrossRef Brunet A, Bonni A, Zigmond MJ, et al. Akt promotes cell survival by phosphorylating and inhibiting a forkhead transcription factor. Cell 1999; 96: 857–68PubMedCrossRef
18.
go back to reference Gnecchi M, He H, Noiseux N, et al. Evidence supporting paracrine hypothesis for Akt-modified mesenchymal stem cell-mediated cardiac protection and functional improvement. FASEB J 2006; 20: 661–9PubMedCrossRef Gnecchi M, He H, Noiseux N, et al. Evidence supporting paracrine hypothesis for Akt-modified mesenchymal stem cell-mediated cardiac protection and functional improvement. FASEB J 2006; 20: 661–9PubMedCrossRef
19.
go back to reference Huff T, Ballweber E, Humeny A, et al. Thymosin β4 serves as a glutaminyl substrate of transglutaminase: labeling with fluorescent dansylcadaverine does not abolish interaction with G-actin. FEBS Lett 1999; 464: 14–20PubMedCrossRef Huff T, Ballweber E, Humeny A, et al. Thymosin β4 serves as a glutaminyl substrate of transglutaminase: labeling with fluorescent dansylcadaverine does not abolish interaction with G-actin. FEBS Lett 1999; 464: 14–20PubMedCrossRef
20.
go back to reference Huff T, Otto AM, Müller CSG, et al. Thymosin β4 is released from human blood platelets and attached by factor XIIIa (transglutaminase) to fibrin and collagen. FASEB J 2002; 16: 691–6PubMedCrossRef Huff T, Otto AM, Müller CSG, et al. Thymosin β4 is released from human blood platelets and attached by factor XIIIa (transglutaminase) to fibrin and collagen. FASEB J 2002; 16: 691–6PubMedCrossRef
21.
go back to reference Alkjaersig N, Fletcher AP, Lewis M, et al. Reduction of coagulation factor XIII concentration in patients with myocardial infarction, cerebral infarction, and other thromboembolic disorders. Thromb Haemost 1977; 38: 863–73PubMed Alkjaersig N, Fletcher AP, Lewis M, et al. Reduction of coagulation factor XIII concentration in patients with myocardial infarction, cerebral infarction, and other thromboembolic disorders. Thromb Haemost 1977; 38: 863–73PubMed
22.
go back to reference Nahrendorf M, Hu K, Frantz S, et al. Factor XIII deficiency causes cardiac rupture, impairs wound healing, and aggravates cardiac remodeling in mice with myocardial infarction. Circulation 2006; 113: 1196–202PubMedCrossRef Nahrendorf M, Hu K, Frantz S, et al. Factor XIII deficiency causes cardiac rupture, impairs wound healing, and aggravates cardiac remodeling in mice with myocardial infarction. Circulation 2006; 113: 1196–202PubMedCrossRef
23.
go back to reference Bereczky Z, Katona E, Muszbek L. Fibrin stabilization (factor XIII), fibrin structure and thrombosis. Pathophysiol Haemost Thromb 2003 Sep–2004 Dec; 33(5–6): 430–7PubMedCrossRef Bereczky Z, Katona E, Muszbek L. Fibrin stabilization (factor XIII), fibrin structure and thrombosis. Pathophysiol Haemost Thromb 2003 Sep–2004 Dec; 33(5–6): 430–7PubMedCrossRef
24.
go back to reference Malinda KM, Goldstein AL, Kleinman HK. Thymosin β4 stimulates directional migration ofhuman umbilical vein endothelial cells. FASEB J 1997; 11: 474–81PubMed Malinda KM, Goldstein AL, Kleinman HK. Thymosin β4 stimulates directional migration ofhuman umbilical vein endothelial cells. FASEB J 1997; 11: 474–81PubMed
25.
go back to reference Grant DS, Kinsella JL, Kibbey MC, et al. Matrigel induces thymosin β4 gene in differentiating endothelial cells. J Cell Sci 1995; 108: 3685–94PubMed Grant DS, Kinsella JL, Kibbey MC, et al. Matrigel induces thymosin β4 gene in differentiating endothelial cells. J Cell Sci 1995; 108: 3685–94PubMed
26.
go back to reference Cha H-J, Jeong M-J, Kleinman HK. Role of thymosin β4 in tumor metastasis and angiogenesis. J Natl Cancer Inst 2003; 95: 1674–80PubMedCrossRef Cha H-J, Jeong M-J, Kleinman HK. Role of thymosin β4 in tumor metastasis and angiogenesis. J Natl Cancer Inst 2003; 95: 1674–80PubMedCrossRef
27.
go back to reference Niu M, Nachmias VT. Increased resistance to apoptosis in cells overexpressing thymosin beta four: a role for focal adhesion kinase pp125FAK. Cell Adhes Commun 2000; 7: 311–20PubMedCrossRef Niu M, Nachmias VT. Increased resistance to apoptosis in cells overexpressing thymosin beta four: a role for focal adhesion kinase pp125FAK. Cell Adhes Commun 2000; 7: 311–20PubMedCrossRef
28.
go back to reference Pradelles P, Frobert Y, Créminon C, et al. Negative regulator of pluripotent hematopoietic stem cell proliferation in human white blood cells and plasma as analysed by enzyme immunoassay. Biochem Biophys Res Commun 1990; 170: 986–93PubMedCrossRef Pradelles P, Frobert Y, Créminon C, et al. Negative regulator of pluripotent hematopoietic stem cell proliferation in human white blood cells and plasma as analysed by enzyme immunoassay. Biochem Biophys Res Commun 1990; 170: 986–93PubMedCrossRef
29.
go back to reference Pradelles P, Frobert Y, Créminon C, et al. Distribution of a negative regulator of haematopoietic stem cell proliferation (AcSDKP) and thymosin β4 in mouse tissues. FEBS Lett 1991; 289: 171–5PubMedCrossRef Pradelles P, Frobert Y, Créminon C, et al. Distribution of a negative regulator of haematopoietic stem cell proliferation (AcSDKP) and thymosin β4 in mouse tissues. FEBS Lett 1991; 289: 171–5PubMedCrossRef
30.
go back to reference Lenfant M, Wdzieczak-Bakala J, Guittet E, et al. Inhibitor of hematopoietic pluripotent stem cell proliferation: purification and determination of its structure. Proc Natl Acad Sci U S A 1989; 86: 779–82PubMedCrossRef Lenfant M, Wdzieczak-Bakala J, Guittet E, et al. Inhibitor of hematopoietic pluripotent stem cell proliferation: purification and determination of its structure. Proc Natl Acad Sci U S A 1989; 86: 779–82PubMedCrossRef
31.
go back to reference Lenfant M, Grillon C, Rieger KJ, et al. Formation of acetyl-Ser-Asp-Lys-Pro, a new regulator of the hematopoietic system, through enzymatic processing of thymosin beta 4. Ann N Y Acad Sci 1991; 628: 115–25PubMedCrossRef Lenfant M, Grillon C, Rieger KJ, et al. Formation of acetyl-Ser-Asp-Lys-Pro, a new regulator of the hematopoietic system, through enzymatic processing of thymosin beta 4. Ann N Y Acad Sci 1991; 628: 115–25PubMedCrossRef
32.
go back to reference Cavasin MA, Rhaleb N-E, Yang X-P, et al. Prolyl oligopeptidase is involved in release of the antifibrotic peptide Ac-SDKP. Hypertension 2004; 43: 1140–5PubMedCrossRef Cavasin MA, Rhaleb N-E, Yang X-P, et al. Prolyl oligopeptidase is involved in release of the antifibrotic peptide Ac-SDKP. Hypertension 2004; 43: 1140–5PubMedCrossRef
33.
go back to reference Azizi M, Rousseau A, Ezan E, et al. Acute angiotensin-converting enzyme inhibition increases the plasma level of the natural stem cell regulator N-acetyl-seryl-aspartyl-lysyl-proline. J Clin Invest 1996; 97: 839–44PubMedCrossRef Azizi M, Rousseau A, Ezan E, et al. Acute angiotensin-converting enzyme inhibition increases the plasma level of the natural stem cell regulator N-acetyl-seryl-aspartyl-lysyl-proline. J Clin Invest 1996; 97: 839–44PubMedCrossRef
34.
go back to reference Azizi M, Ezan E, Nicolet L, et al. High plasma level of N-acetyl-seryl-aspartyl-lysyl-proline: a new marker of chronic angiotensin-converting enzyme inhibition. Hypertension 1997; 30: 1015–9PubMedCrossRef Azizi M, Ezan E, Nicolet L, et al. High plasma level of N-acetyl-seryl-aspartyl-lysyl-proline: a new marker of chronic angiotensin-converting enzyme inhibition. Hypertension 1997; 30: 1015–9PubMedCrossRef
35.
go back to reference Rhaleb N-E, Peng H, Harding P, et al. Effect of N-acetyl-seryl-aspartyl-lysyl-proline on DNA and collagen synthesis in rat cardiac fibroblasts. Hypertension 2001; 37: 827–32PubMedCrossRef Rhaleb N-E, Peng H, Harding P, et al. Effect of N-acetyl-seryl-aspartyl-lysyl-proline on DNA and collagen synthesis in rat cardiac fibroblasts. Hypertension 2001; 37: 827–32PubMedCrossRef
36.
go back to reference Peng H, Carretero OA, Brigstock DR, et al. Ac-SDKP reverses cardiac fibrosis in rats with renovascular hypertension. Hypertension 2003; 42: 1164–70PubMedCrossRef Peng H, Carretero OA, Brigstock DR, et al. Ac-SDKP reverses cardiac fibrosis in rats with renovascular hypertension. Hypertension 2003; 42: 1164–70PubMedCrossRef
37.
go back to reference Rhaleb N-E, Peng H, Yang X-P, et al. Long-term effect of N-acetyl-seryl-aspartyl-lysyl-proline on left ventricular collagen deposition in rats with 2-kidney, 1-clip hypertension. Circulation 2001; 103: 3136–41PubMedCrossRef Rhaleb N-E, Peng H, Yang X-P, et al. Long-term effect of N-acetyl-seryl-aspartyl-lysyl-proline on left ventricular collagen deposition in rats with 2-kidney, 1-clip hypertension. Circulation 2001; 103: 3136–41PubMedCrossRef
38.
go back to reference Yang F, Yang X-P, Liu Y-H, et al. Ac-SDKP reverses inflammation and fibrosis in rats with heart failure after myocardial infarction. Hypertension 2004; 43: 229–36PubMedCrossRef Yang F, Yang X-P, Liu Y-H, et al. Ac-SDKP reverses inflammation and fibrosis in rats with heart failure after myocardial infarction. Hypertension 2004; 43: 229–36PubMedCrossRef
39.
go back to reference Peng H, Carretero OA, Vuljaj N, et al. Angiotensin-converting enzyme inhibitors: a new mechanism of action. Circulation 2005; 112: 2436–45PubMedCrossRef Peng H, Carretero OA, Vuljaj N, et al. Angiotensin-converting enzyme inhibitors: a new mechanism of action. Circulation 2005; 112: 2436–45PubMedCrossRef
40.
go back to reference Miller EJ, Gay S. Collagen structure and function. In: Cohen IK, Diegelmann RF, Lindblad WJ, editors. Wound healing: biochemical and clinical aspects. Philadelphia (PA): WB Saunders, 1992: 130–51 Miller EJ, Gay S. Collagen structure and function. In: Cohen IK, Diegelmann RF, Lindblad WJ, editors. Wound healing: biochemical and clinical aspects. Philadelphia (PA): WB Saunders, 1992: 130–51
41.
go back to reference Smith-Mungo LI, Kagan HM. Lysyl oxidase: properties, regulation and multiple functions in biology. Matrix Biol 1998; 16: 387–98PubMedCrossRef Smith-Mungo LI, Kagan HM. Lysyl oxidase: properties, regulation and multiple functions in biology. Matrix Biol 1998; 16: 387–98PubMedCrossRef
42.
go back to reference Mäki JM, Sormunen R, Lippo S, et al. Lysyl oxidase is essential for normal development and function of the respiratory system and for the integrity of elastic and collagen fibers in various tissues. Am J Pathol 2005; 167: 927–36PubMedCrossRef Mäki JM, Sormunen R, Lippo S, et al. Lysyl oxidase is essential for normal development and function of the respiratory system and for the integrity of elastic and collagen fibers in various tissues. Am J Pathol 2005; 167: 927–36PubMedCrossRef
43.
go back to reference Liu J-M, Lawrence F, Kovacevic M, et al. The tetrapeptide AcSDKP, an inhibitor of primitive hematopoietic cell proliferation, induces angiogenesis in vitro and in vivo. Blood 2003; 101: 3014–20PubMedCrossRef Liu J-M, Lawrence F, Kovacevic M, et al. The tetrapeptide AcSDKP, an inhibitor of primitive hematopoietic cell proliferation, induces angiogenesis in vitro and in vivo. Blood 2003; 101: 3014–20PubMedCrossRef
44.
go back to reference Irazusta J, Silveira PF, Gil J, et al. Effects of hydrosaline treatments on prolyl endopeptidase activity in rat tissues. Regul Pept 2001; 101: 141–7PubMedCrossRef Irazusta J, Silveira PF, Gil J, et al. Effects of hydrosaline treatments on prolyl endopeptidase activity in rat tissues. Regul Pept 2001; 101: 141–7PubMedCrossRef
46.
go back to reference Kimura A, Matsui H, Takahashi T. Expression and localization of prolyl oligopeptidase in mouse testis and its possible involvement in sperm motility. Zoolog Sci 2002; 19: 93–102PubMedCrossRef Kimura A, Matsui H, Takahashi T. Expression and localization of prolyl oligopeptidase in mouse testis and its possible involvement in sperm motility. Zoolog Sci 2002; 19: 93–102PubMedCrossRef
47.
go back to reference Goossens F, De Meester I, Vanhoof G, et al. Distribution of prolyl oligopeptidase in human peripheral tissues and body fluids. Eur J Clin Chem Clin Biochem 1996; 34: 17–22PubMed Goossens F, De Meester I, Vanhoof G, et al. Distribution of prolyl oligopeptidase in human peripheral tissues and body fluids. Eur J Clin Chem Clin Biochem 1996; 34: 17–22PubMed
48.
go back to reference Wang D, Carretero OA, Yang X-Y, et al. N-acetyl-seryl-aspartyl-lysyl-proline stimulates angiogenesis in vitro and in vivo. Am J Physiol Heart Circ Physiol 2004; 287: H2099–105PubMedCrossRef Wang D, Carretero OA, Yang X-Y, et al. N-acetyl-seryl-aspartyl-lysyl-proline stimulates angiogenesis in vitro and in vivo. Am J Physiol Heart Circ Physiol 2004; 287: H2099–105PubMedCrossRef
49.
go back to reference Omata M, Taniguchi H, Koya D, et al. N-acetyl-seryl-aspartyl-lysyl-proline ameliorates the progression of renal dysfunction and fibrosis in WKY rats with established anti-glomerular basement membrane nephritis. J Am Soc Nephrol 2006; 17: 674–85PubMedCrossRef Omata M, Taniguchi H, Koya D, et al. N-acetyl-seryl-aspartyl-lysyl-proline ameliorates the progression of renal dysfunction and fibrosis in WKY rats with established anti-glomerular basement membrane nephritis. J Am Soc Nephrol 2006; 17: 674–85PubMedCrossRef
50.
go back to reference Koutrafouri V, Leondiadis L, Avgoustakis K, et al. Effect of thymosin peptides on the chick chorioallantoic membrane angiogenesis model. Biochim Biophys Acta 2001; 1568: 60–6PubMedCrossRef Koutrafouri V, Leondiadis L, Avgoustakis K, et al. Effect of thymosin peptides on the chick chorioallantoic membrane angiogenesis model. Biochim Biophys Acta 2001; 1568: 60–6PubMedCrossRef
51.
go back to reference Xu J, Carretero OA, Rhaleb N-E, et al. Effect of Ac-SDKP on cardiac rupture and early remodeling after myocardial infarction in mice [abstract]. Circulation 2004; 110: III–27 Xu J, Carretero OA, Rhaleb N-E, et al. Effect of Ac-SDKP on cardiac rupture and early remodeling after myocardial infarction in mice [abstract]. Circulation 2004; 110: III–27
52.
go back to reference Waeckel L, Bignon J, Liu J-M, et al. Tetrapeptide AcSDKP induces postischemic neovascularization through monocyte chemoattractant protein-1 signaling. Arterioscler Thromb Vasc Biol 2006; 26: 773–9PubMedCrossRef Waeckel L, Bignon J, Liu J-M, et al. Tetrapeptide AcSDKP induces postischemic neovascularization through monocyte chemoattractant protein-1 signaling. Arterioscler Thromb Vasc Biol 2006; 26: 773–9PubMedCrossRef
53.
go back to reference Sosne G, Christopherson PL, Barrett RP, et al. Thymosin-β4 modulates corneal matrix metalloproteinase levels and polymorphonuclear cell infiltration after alkali injury. Invest Ophthalmol Vis Sci 2005; 46: 2388–95PubMedCrossRef Sosne G, Christopherson PL, Barrett RP, et al. Thymosin-β4 modulates corneal matrix metalloproteinase levels and polymorphonuclear cell infiltration after alkali injury. Invest Ophthalmol Vis Sci 2005; 46: 2388–95PubMedCrossRef
54.
go back to reference Pokharel S, Rasoul S, Roks AJM, et al. N-acetyl-Ser-Asp-Lys-Pro inhibits phosphorylation of Smad2 in cardiac fibroblasts. Hypertension 2002; 40: 155–61PubMedCrossRef Pokharel S, Rasoul S, Roks AJM, et al. N-acetyl-Ser-Asp-Lys-Pro inhibits phosphorylation of Smad2 in cardiac fibroblasts. Hypertension 2002; 40: 155–61PubMedCrossRef
Metadata
Title
Therapeutic Potential of Thymosin-β4 and its Derivative N-acetyl-seryl-aspartyl-lysyl-proline (Ac-SDKP) in Cardiac Healing After Infarction
Author
Dr Maria A. Cavasin
Publication date
01-09-2006
Publisher
Springer International Publishing
Published in
American Journal of Cardiovascular Drugs / Issue 5/2006
Print ISSN: 1175-3277
Electronic ISSN: 1179-187X
DOI
https://doi.org/10.2165/00129784-200606050-00003

Other articles of this Issue 5/2006

American Journal of Cardiovascular Drugs 5/2006 Go to the issue

Review Article

The ASCOT Trial