钙调神经磷酸酶信号通路在心肌肥厚和心衰发生机制中的研究
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
研究背景:
     心力衰竭是不同病因所致的心脏病发展的最终结局。目前对心力衰竭的防治虽取得了不少进展,但其患病率和死亡率仍居高不下,因而进一步加强心力衰竭的防治研究有着重大的意义。心肌肥厚是临床上发生心力衰竭的前期表现,是心肌为了满足不断增加的心脏做功需要而发生的代偿性变化,这种变化在短时间内可维持或增加心输出量。但心肌肥厚也会增加心肌耗氧量,降低冠状动脉血流储备,长期的供需失衡将会使“代偿性”肥厚逐渐发展为“失代偿性”肥厚,如果再同时合并心肌纤维化和细胞凋亡,最终将导致心功能下降而出现心衰的临床表现。因此,探讨心肌肥厚发生机制以及如何进展为心力衰竭非常重要,可能由此而发现新的心力衰竭防治措施。
     心肌肥厚的病理变化包括基因转录和蛋白合成增加,以及肌原纤维重新组装,多数合并胚胎基因表达增加,包括利钠肽和胎儿收缩蛋白基因。利钠肽基因的出现是所有哺乳动物心肌肥厚的特征,也是临床上判断疾病严重程度的预测指标。迄今为止,已发现了许多可引起心肌肥厚的病理生理刺激因子,如机械应力、G-蛋白偶联受体(GPCR)激动剂(包括内皮素-1、人尿压素Ⅱ、血管紧张素-Ⅱ)、细胞因子和生长因子。这些刺激因子涉及到许多细胞内信号传导通路,包括丝裂原活化蛋白激酶(MAPK)、蛋白激酶C(PKC)、磷脂酰肌醇3-激酶(PI3-K)-Akt(又称蛋白激酶B)、Janus激酶-信号传导与转录活化因子(JAK-STAT)和白介素6-白介素6受体-糖蛋白130(IL6-IL6R-gp130)。近年来,钙调神经磷酸酶(Calcineurin)作为Ca~(2+)-介导造成心肌肥厚的重要调
Background: Heart failure is an important and growing public health problem and the cause of substantial morbidity and mortality. Hypertrophy is an early event in the clinical course of heart failure. In response to increased cardiac work demand, the heart increases muscle mass thereby providing temporary maintenance of cardiac output. In short term hypertrophy maintains or increases cardiac output. So it can be viewed as a "beneficial" compensatory response. However, hypertrophy also increases oxygen consumption and reduces coronary blood-flow reserve. This supply-demand mismatch may lead the patient to angina pectoris, MI, arrhythmia or sudden death. In addition, during the process of hypertrophy, cardiac fibrosis and apoptosis may occur simultaneously. All these co-existing pathologies mediate the eventual decline in myocardial function and lead to congestive heart failure. So, the investigation of mechanisms for cardiac hypertrophy and its progression to heart failure is very important, as this may lead to the development of new therapeutic modalities and better guidelines for the prevention of cardiac disease.Pathological changes resulting in cardiac hypertrophy include the increase of gene transcription, protein synthesis, and myofibril assembly. In most forms of cardiac hypertrophy, there is an increase in the expression of embryonic genes, including the genes of natriuretic peptides and fetal contractile proteins. The induction of the natriuretic peptide genes is a feature of hypertrophy in all-mammalian species and is a prognostic indicator for clinical severity of disease. So far, numerous pathophysiological stimuli have been identified that can induce cardiac hypertrophy, for example mechanical stress, G-protein coupled receptor (GPCR) agonists (including endothelin-1, human urotensin Ⅱ, and angiotensin-Ⅱ), cytokines, and growth factors. Many intracellular signalling pathways have also been implicated; including the mitogen-activated protein kinases (MAPKs), protein
    kinase C (PKC), phosphatidyl inositol 3 kinase (PI3 K) -Akt, Janus kinase - Signal Transducers and Activators of Transcription (JAK-STAT), interleukin 6 -interleukin 6 receptor - glycoprotein 130 (IL6-IL6R-gpl30). Recently, calcineurin has attracted great attention as a mediator of Ca2+ -induced cardiac hypertrophy. Over expression of constitutively active mutants of calcineurin and of its downstream transcription factor, nuclear factor of activated T-cells (NFAT3) induced marked cardiac hypertrophy in transgenic mice. The calcineurin inhibitors cyclosporin A and FK506 suppressed phenylephrine- and angiotensin II -induced cardiomyocyte hypertrophy in vitro. All these proved that calcineurin signalling pathway is important for cardiac hypertrophy and heart failure.Our previous study used a tissue bath method to investigate the role of G-protein coupled receptor (GPCR) agonists, including endothelin - 1 (ET-1), angiotensin - II (Ang-II), and human urotensin - II (hUII), on calcineurin activity in human heart. Results showed that all these agonists can significantly increased calcineurin activity. This raised a question, "what's the mechanism by which calcineurin activity was increased?"Aim: The purpose of this study was to investigate the mechanism by which GPCR agonists increase calcineurin activity. Before study we made three working hypothesises that might explain increased calcineurin activity following agonist stimulation. First one: GPCR agonists increase calcineurin activity by increase its protein expression; Second one: after activated by GPCR agonists, PKC phosphorylate some proteins, which also were substrates of calcienurin, increase the calcineurin-mediate free phosphates formation. Thereby increase "calcineurin activity"; the last one: after stimulated by GPCR agonists, calcineurin can be limited proteolysed into constitutively active form, and no need Ca2+/calmodulin for its activity. Calpain is a strong candidate of the enzymes responsible for the post-translational modification of calcineurin.Methods and Results: We examined in the study the possible mechanism by
    which GPCR agonists stimulate calcineurin activity in human heart.Patient Population: Tissue from left and right ventricles was obtained from terminal heart failure patients (n=12) undergoing heart transplantation. They were diagnosed with ischaemic heart disease (IHD) (n=5), idiopathic dilated cardiomyopathy (IDC) (n=3), familial dilated cardiomyopathy (n=l), single ventricle with Damus connection (n=l), aortic valve disease (AVD) (n=l), and Becker's syndrome (n=l). All patients had left ventricular dysfunction and elevated left ventricular end diastolic pressure. In some patients, the increased LV pressure was transmitted to the right side of the heart (mean right ventricular systolic pressure 49.5±3.5mmHg), and all patients had right ventricular dysfunction as determined by echocardiogram.The donor hearts that served as the "controls" (n=5) were obtained from subjects who had died from non-cardiac disease related causes, for example victims of vehicle accidents. Their clinical data were unclear.Methods: Several molecular biological technics, such as Western blot, immunohistochemistry, cell culture, zymography, and BioMol calcineurin activity assay, et al were employed in the study.1. Western Blot: In order to know whether calcineurin activity was infected by its protein express, and whether protein expression was related to its distribution, state of heart function, aetiology, and agonists stimulation, we using Western blot method compared calcineurin protein expression between left and right ventricle, failing right ventricle and "donor" right ventricle, right ventricle before and after agonists stimulation in Tissue bath experiment, right ventricle of low and high basal calcineurin activity determined in Tissue bath exprement.2. Immunohistochemistry: Calcineurin is widely distributed in various mammalian tissues. Its concentration in brain is 10-20 times greater than in other tissues and constitutes about 1% of the total protein concentration. It has been learned more and more about the distribution of calcineurin in nerves system, such as 50-70% of calcineurin is bound to membranous or cytoskeletal elements, almost exclusively neuronal distribution of calcineurin in the brain of
    different species, CnAa is located in the nuclei while CnAfi is located in the cytoplasm, et al. Compared to the neuronal system, very little is known about the distribution of calcineurin in heart. Heart contains a heterogeneous population of cell types, including cardiomyocytes, fibroblasts, smooth muscle cells, epicardial mesothelial cells, endocardial and vascular endothelial cells, and inflammatory cells. In which cell type does calcineurin expressed? We'll using immunohistochemistry technic to investigate about these.3. Calcineurin activity assay: By calcineurin activity assay to explore the contribution of PKC and calpain to calcineurin activation. Different isoforms of recombinant PKC, calcineurin, and purified calpain were used in this study. To specify PKC and calpain action, we use okadaic acid (OA) to inhibit other phosphatases, such as PP1 and PP2A. Calpain inhibitor leupeptin was used to prove calpain function. In addition, we also investigated the calcium dependence of calpain activation.4. Zymography: Zymography was performed to see the calpain activity in human heart.5. Cell Culture: Cardiac fibroblasts were grown from abdominal aortic artery. Stimulated by ET-1 and hUII respectively in different time. Observe the changes of calcineurin activity and protein fragments.Results:1. Calcineurin activity was independent of protein expression. And protein expression does not influenced by aetiology, ventricle location, and short time agonist stimulation.2. The current study showed that calcineurin was expressed in cardiac myocytes and fibroblasts, a finding consistent with the reported role of calcineurin in myocardial hypertrophy and fibrosis. Staining was also detected in epicardial mesothelial cells, although the role of calcineurin at the surface of the heart is not known. No staining was found in vascular endothelial or smooth muscle cells.3. Different isoforms of PKC have different effect on calcineurin activity. PKCe
    really can increase calcineurin activity by increase calcineurin substrates. While in samples that were pre-incubated with PKCaPy, a non-significant increase trend for basal (no PKC) calcineurin-dependent free phosphate formation was observed.4. Calpain expressed in human ventricle.5. Both PKC and calcineurin were substrate of calpain. Requirement of calpain I and calpain II for Ca2+ was different, the truncation products of calcineurin were not same for calpain I and calpain II. An apparent molecular mass of 45 KDa was generated in the calpain I treated sample, whereas smaller cleavage fragments were detected using calpain II. After proteolysis, calcineurin becomes constitutively active form, which no need Ca2+/calmodulin for activation.6. The ability of GPCR agonists to stimulate calpain-dependent activation of calcineurin was investigated. However, protein concentration was too low to detect calcineurin bands.Results show that: 1. calcineurin expression was independent of calcineurin activity; 2. PKCs really increase "calcineurin activity" by increase calcineurin substrates; 3. Both calcineurin and PKC are substrates of calpain. After limited proteolysis, calcineurin activity can be increased significantly, which no need Ca2+/calmodulin for its activation.Conclusion: The mechanism of GPCR agonists increase calcineurin activity was related to PKC and calpain, and was independent of calcineurin expression. After stimulation by GPCR agonists, PKC increase calcineurin activity by increase calcineurin substrates; while calpain by limited proteolysis of calcineurin. After proteolysis, calcineurin was no need Ca2+/calmodulin for its activation.New points of this study: Recent years, the knowledge about Ca2+_ calcineurin _ NFAT signalling pathway in cardiac hypertrophy and heart failure has been learned more and more. However, about the mechanism by which calcineurin
    was activated was still unknown. This study is the first attempt to explain it. These hypothesises were made before investigation. Two of them were proved by this study, and one got negative result. Anyway, together all the results, it will be a good contribution to complete the mechanism of hypertrophy and heart failure. We may provide theory basis for prevent cardiac hypertrophy.
引文
1. Chien KR, Zhu H, Knowlton KU, Miller-Hance W, van-Bilsen M, O'Brien TX, Evans SM. Transcriptional regulation during cardiac growth and development. Annu Rev Physiol. 1993; 55: 77-95.
    2. Pelliccia A, Maron BJ. Outer limits of the athlete's heart, the effect of gender, and relevance to the differential diagnosis with primary cardiac diseases. Cardiol Clin. 1997; 15: 381-96.
    3. Oakley D. General cardiology: The athlete's heart. Heart. 2001; 86: 722-6.
    4. Pluim BM, Zwinderman AH, van der Laarse A, van der Wall EE. The athlete's heart. A meta-analysis of cardiac structure and function. Circulation. 2000; 101: 336-44.
    5. Devereux RB, Casale PN, Hammond IW, Savage DD, Alderman MH, Campo E, Alonso DR, Laragh JH. Echocardiographic detection of pressure-overload left ventricular hypertrophy: effect of criteria and patient population. J Clin Hypertens. 1987; 3: 66-78.
    6. Maron BJ, Epstein SE, Roberts WC. Causes of sudden death in competitive athletes. J Am Coll Cardiol. 1986; 7: 204-14.
    7. Maron BJ, Gardin JM, Flack JM, Gidding SS, Kurosaki TT, Bild DE. Prevalence of hypertrophic cardiomyopathy in a general population of young adults. Echocardiographic analysis of 4111 subjects in the CARDIA Study. Coronary Artery Risk Development in (Young) Adults. Circulation. 1995; 92: 785-9.
    8. Smith SH, Bishop SP. Regional myocyte size in compensated right ventricular hypertrophy in the ferret. J Mol Cell Cardiol. 1985; 17: 1005-11.
    9. Campbell SE, Korecky B, Rakusan K. Remodeling of myocyte dimensions in hypertrophic and atrophic rat hearts. Circ Res. 1991; 68: 984-96.
    10. Gerdes AM, Moore JA, Hines JM. Regional changes in myocyte size and number in propranolol-treated hyperthyroid rats. Lab Invest. 1987; 57: 708-13.
    11. Koyanagi S, Eastham C, Marcus ML. Effects of chronic hypertension and left ventricular hypertrophy on the incidence of sudden cardiac death after coronary artery occlusion in conscious dogs. Circulation. 1982;65:1192-7.
    12. Koyanagi S, Eastham CL, Harrison DG, Marcus ML. Increased size of myocardial infarction in dogs with chronic hypertension and left ventricular hypertrophy. Circ Res. 1982;50:55-62.
    13. Levy D, Anderson KM, Savage DD, Balkus SA, Kannel WB, Castelli WP. Risk of ventricular arrhythmias in left ventricular hypertrophy: the Framingham Heart Study. Am J Cardiol. 1987;60:560-5.
    14. McLenachan JM, Henderson E, Morris KI, Dargie HJ. Ventricular arrhythmias in patients with hypertensive left ventricular hypertrophy. N Engl J Med. 1987;317:787-92.
    15. Paradis P, Dali-Youcef N, Paradis FW, Thibault G, Nemer M. Overexpression of angiotensin II type I receptor in cardiomyocytes induces cardiac hypertrophy and remodeling. Proc Natl Acad Sci U S A. 2000;97:931-6.
    16. D'Angelo DD, Sakata Y, Lorenz JN, Boivin GP, Walsh RA, Liggett SB, Dorn GW, 2nd. Transgenic Galphaq overexpression induces cardiac contractile failure in mice. In: Proc Natl Acad Sci U S A ; 1997:8121-6.
    17. Milano CA, Dolber PC, Rockman HA, Bond RA, Venable ME, Allen LF, Lefkowitz RJ. Myocardial expression of a constitutively active alpha 1B-adrenergic receptor in transgenic mice induces cardiac hypertrophy. Proc Natl Acad Sci U S A. 1994;91:10109-13.
    18. Ito H, Hirata Y, Hiroe M, Tsujino M, Adachi S, Takamoto T, Nitta M, Taniguchi K, Marumo F. Endothelin-1 induces hypertrophy with enhanced expression of muscle-specific genes in cultured neonatal rat cardiomyocytes. Circ Res. 1991;69:209-15.
    19. Dostal DE, Hunt RA, Kule CE, Bhat GJ, Karoor V, McWhinney CD, Baker KM. Molecular mechanisms of angiotensin II in modulating cardiac function: intracardiac effects and signal transduction pathways. J Mol Cell Cardiol. 1997; 29: 2893-902.
    20. Tzanidis A, Hannan RD, Thomas WG, Onan D, Autelitano DJ, See F, Kelly DJ, Gilbert RE, Krum H. Direct actions of urotensin Ⅱ on the heart: implications for cardiac fibrosis and hypertrophy. Circ Res. 2003; 93: 246-53.
    21. Onan D, Pipolo L, Yang E, Hannan RD, Thomas WG. Urotensin Ⅱ promotes hypertrophy of cardiac myocytes via mitogen-activated protein kinases. Mol Endocrinol. 2004; 18: 2344-54.
    22. Braunwald E, Bristow MR. Congestive heart failure: fifty years of progress. Circulation. 2000; 102:Ⅳ 14-23.
    23. Sakai S, Miyauchi T, Kobayashi M, Yamaguchi I, Goto K, Sugishita Y. Inhibition of myocardial endothelin pathway improves long-term survival in heart failure. Nature. 1996; 384: 353-5.
    24. Yoshida H, Takahashi M, Tanonaka K, Maki T, Nasa Y, Takeo S. Effects of ACE inhibition and angiotensin Ⅱ type 1 receptor blockade on cardiac function and G proteins in rats with chronic heart failure. Br J Pharmacol. 2001; 134: 150-60.
    25. Russell FD, Molenaar P. Investigation of signaling pathways that mediate the inotropic effect of urotensin-Ⅱ in human heart. Cardiovasc Res. 2004; 63: 673-81.
    26. Maguire JJ, Davenport AP. Is urotensin-Ⅱ the new endothelin? Br J Pharmacol. 2002; 137: 579-88.
    27. Ito H, Hirata Y, Hiroe M, Tsujino M, Adachi S, Takamoto T, Nitta M, Taniguchi K, Marumo F. Endothelin-1 induces hypertrophy with enhanced expression of muscle-specific genes in cultured neonatal rat cardiomyocytes. In: Circ Res; 1991: 209-15.
    28. Sadoshima J, Xu Y, Slayter HS, Izumo S. Autocrine release of angiotensin Ⅱ mediates stretch-induced hypertrophy of cardiac myocytes in vitro. Cell. 1993; 75: 977-84.
    29. Zou Y, Nagai R, Yamazaki T. Urotensin Ⅱ induces hypertrophic responses in cultured cardiomyocytes from neonatal rats. FEBS Lett. 2001;508:57-60.
    30. Sadoshima J, Izumo S. The cellular and molecular response of cardiac myocytes to mechanical stress. Annu Rev Physiol. 1997;59:551-71.
    31. de Gasparo M, Catt KJ, Inagami T, Wright JW, Unger T. International union of pharmacology. XXIII. The angiotensin II receptors. Pharmacol Rev. 2000;52:415-72.
    32. Thomas WG, Brandenburger Y, Autelitano DJ, Pham T, Qian H, Hannan RD. Adenoviral-directed expression of the type 1A angiotensin receptor promotes cardiomyocyte hypertrophy via transactivation of the epidermal growth factor receptor. Circ Res. 2002;90:135-42.
    33. Asakura M, Kitakaze M, Takashima S, Liao Y, Ishikura F, Yoshinaka T, Ohmoto H, Node K, Yoshino K, Ishiguro H, Asanuma H, Sanada S, Matsumura Y, Takeda H, Beppu S, Tada M, Hori M, Higashiyama S. Cardiac hypertrophy is inhibited by antagonism of ADAM12 processing of HB-EGF: metalloproteinase inhibitors as a new therapy. Nat Med. 2002;8:35-40.
    34. Crone SA, Zhao YY, Fan L, Gu Y, Minamisawa S, Liu Y, Peterson KL, Chen J, Kahn R, Condorelli G, Ross J, Jr., Chien KR, Lee KF. ErbB2 is essential in the prevention of dilated cardiomyopathy. Nat Med. 2002;8:459-65.
    35. Rybin VO, Steinberg SF. Protein kinase C isoform expression and regulation in the developing rat heart. Circ Res. 1994;74:299-309.
    36. Puceat M, Hilal-Dandan R, Strulovici B, Brunton LL, Brown JH. Differential regulation of protein kinase C isoforms in isolated neonatal and adult rat cardiomyocytes. J Biol Chem. 1994;269:16938-44.
    37. Clerk A, Bogoyevitch MA, Anderson MB, Sugden PH. Differential activation of protein kinase C isoforms by endothelin-1 and phenylephrine and subsequent stimulation of p42 and p44 mitogen-activated protein kinases in ventricular myocytes cultured from neonatal rat hearts. J Biol Chem. 1994;269:32848-57.
    38. Dunnmon PM, Iwaki K, Henderson SA, Sen A, Chien KR. Phorbol esters induce immediate-early genes and activate cardiac gene transcription in neonatal rat myocardial cells. J Mol Cell Cardiol. 1990;22:901-10.
    39. Wakasaki H, Koya D, Schoen FJ, Jirousek MR, Ways DK, Hoit BD, Walsh RA, King GL. Targeted overexpression of protein kinase C beta2 isoform in myocardium causes cardiomyopathy. Proc Natl Acad Sci U S A. 1997;94:9320-5.
    40. Bowman JC, Steinberg SF, Jiang T, Geenen DL, Fishman GI, Buttrick PM. Expression of protein kinase C beta in the heart causes hypertrophy in adult mice and sudden death in neonates. J Clin Invest. 1997;100:2189-95.
    41. Takeishi Y, Ping P, Bolli R, Kirkpatrick DL, Hoit BD, Walsh RA. Transgenic overexpression of constitutively active protein kinase C epsilon causes concentric cardiac hypertrophy. Circ Res. 2000;86:1218-23.
    42. Mitchell RD, Simmerman HK, Jones LR. Ca2+ binding effects on protein conformation and protein interactions of canine cardiac calsequestrin. J Biol Chem. 1988;263:1376-81.
    43. Ikemoto N, Ronjat M, Meszaros LG, Koshita M. Postulated role of calsequestrin in the regulation of calcium release from sarcoplasmic reticulum. Biochemistry. 1989;28:6764-71.
    44. Zhang L, Kelley J, Schmeisser G, Kobayashi YM, Jones LR. Complex formation between junctin, triadin, calsequestrin, and the ryanodine receptor. Proteins of the cardiac junctional sarcoplasmic reticulum membrane. J Biol Chem. 1997;272:23389-97.
    45. Knollmann BC, Knollmann-Ritschel BE, Weissman NJ, Jones LR, Morad M. Remodelling of ionic currents in hypertrophied and failing hearts of transgenic mice overexpressing calsequestrin. J Physiol. 2000;525 Pt 2:483-98.
    46. Linck B, Boknik P, Huke S, Kirchhefer U, Knapp J, Luss H, Muller FU, Neumann J, Tanriseven Z, Vahlensieck U, Baba HA, Jones LR, Philipson KD, Schmitz W. Functional properties of transgenic mouse hearts overexpressing both calsequestrin and the Na(+)-Ca(2+) exchanger. J Pharmacol Exp Ther. 2000;294:648-57.
    47. Ihara Y, Suzuki YJ, Kitta K, Jones LR, Ikeda T. Modulation of gene expression in transgenic mouse hearts overexpressing calsequestrin. Cell Calcium. 2002;32:21-9.
    48. Bers DM, Bridge JH. Relaxation of rabbit ventricular muscle by Na-Ca exchange and sarcoplasmic reticulum calcium pump. Ryanodine and voltage sensitivity. Circ Res. 1989;65:334-42.
    49. Pott C, Goldhaber JI, Philipson KD. Genetic manipulation of cardiac Na+/Ca2+ exchange expression. Biochem Biophys Res Commun. 2004;322:1336-40.
    50. Komuro I, Wenninger KE, Philipson KD, Izumo S. Molecular cloning and characterization of the human cardiac Na+/Ca2+ exchanger cDNA. Proc NatlAcadSci USA. 1992;89:4769-73.
    51. Kiss E, Ball NA, Kranias EG, Walsh RA. Differential changes in cardiac phospholamban and sarcoplasmic reticular Ca(2+)-ATPase protein levels. Effects on Ca2+ transport and mechanics in compensated pressure-overload hypertrophy and congestive heart failure. Circ Res. 1995;77:759-64.
    52. Nagai R, Zarain-Herzberg A, Brandl CJ, Fujii J, Tada M, MacLennan DH, Alpert NR, Periasamy M. Regulation of myocardial Ca2+-ATPase and phospholamban mRNA expression in response to pressure overload and thyroid hormone. Proc Natl Acad Sci USA. 1989;86:2966-70.
    53. Sugden PH. Signaling in myocardial hypertrophy: life after calcineurin? Circ Res. 1999;84:633-46.
    54. Clerk A, Sugden PH. Cell stress-induced phosphorylation of ATF2 and c-Jun transcription factors in rat ventricular myocytes. Biochem J. 1997;325 (Pt3):801-10.
    55. Clerk A, Sugden PH. Phosphorylation c-Jun and ATF2 in ventricular myocytes by endothelin and phenylephrine. Biochem Soc Trans. 1997;25:222S.
    56. Han J, Jiang Y, Li Z, Kravchenko VV, Ulevitch RJ. Activation of the transcription factor MEF2C by the MAP kinase p38 in inflammation. Nature. 1997;386:296-9.
    57. Dhand R, Hiles I, Panayotou G, Roche S, Fry MJ, Gout I, Totty NF, Truong O, Vicendo P, Yonezawa K, et al. PI 3-kinase is a dual specificity enzyme: autoregulation by an intrinsic protein-serine kinase activity. Embo J. 1994;13:522-33.
    58. Oudit GY, Sun H, Kerfant BG, Crackower MA, Penninger JM, Backx PH. The role of phosphoinositide-3 kinase and PTEN in cardiovascular physiology and disease. J Mol Cell Cardiol. 2004;37:449-71.
    59. Shioi T, Kang PM, Douglas PS, Hampe J, Yballe CM, Lawitts J, Cantley LC, Izumo S. The conserved phosphoinositide 3-kinase pathway determines heart size in mice. Embo J. 2000; 19:2537-48.
    60. Taga T, Kishimoto T. Gpl30 and the interleukin-6 family of cytokines. Annu Rev Immunol. 1997;15:797-819.
    61. Eiken HG, Oie E, Damas JK, Yndestad A, Bjerkeli V, Aass H, Simonsen S, Geiran OR, Tonnessen T, Christensen G, Froland SS, Gullestad L, Attramadal H, Aukrust P. Myocardial gene expression of leukaemia inhibitory factor, interleukin-6 and glycoprotein 130 in end-stage human heart failure. Eur J Clin Invest. 2001 ;31:389-97.
    62. Hirota H, Yoshida K, Kishimoto T, Taga T. Continuous activation of gpl30, a signal-transducing receptor component for interleukin 6-related cytokines, causes myocardial hypertrophy in mice. Proc Natl Acad Sci U S A . 1995;92:4862-6.
    63. Yoshida K, Taga T, Saito M, Suematsu S, Kumanogoh A, Tanaka T, Fujiwara H, Hirata M, Yamagami T, Nakahata T, Hirabayashi T, Yoneda Y, Tanaka K, Wang WZ, Mori C, Shiota K, Yoshida N, Kishimoto T. Targeted disruption of gp130, a common signal transducer for the interleukin 6 family of cytokines, leads to myocardial and hematological disorders. Proc Natl AcadSci USA. 1996;93:407-11.
    64. Betz UA, Bloch W, van den Broek M, Yoshida K, Taga T, Kishimoto T, Addicks K, Rajewsky K, Muller W. Postnatally induced inactivation of gp130 in mice results in neurological, cardiac, hematopoietic,immunological, hepatic, and pulmonary defects. J Exp Med. 1998;188:1955-65.
    65. Stemmer PM, Klee CB. Dual calcium ion regulation of calcineurin by calmodulin and calcineurin B. Biochemistry. 1994;33:6859-66.
    66. Bueno OF, van Rooij E, Molkentin JD, Doevendans PA, De Windt LJ. Calcineurin and hypertrophic heart disease: novel insights and remaining questions. Cardiovasc Res. 2002;53:806-21.
    67. Muramatsu T, Giri PR, Higuchi S, Kincaid RL. Molecular cloning of a calmodulin-dependent phosphatase from murine testis: identification of a developmentally expressed nonneural isoenzyme. Proc Natl Acad Sci U S A. 1992;89:529-33.
    68. Hayden-Martinez K, Kane LP, Hedrick SM. Effects of a constitutively active form of calcineurin on T cell activation and thymic selection. J Immunol. 2000;165:3713-21.
    69. Yakel JL. Calcineurin regulation of synaptic function: from ion channels to transmitter release and gene transcription. Trends Pharmacol Sci. 1997;18:124-34.
    70. Lautermilch NJ, Spitzer NC. Regulation of calcineurin by growth cone calcium waves controls neurite extension. J Neurosci. 2000;20:315-25.
    71. de la Pompa JL, Timmerman LA, Takimoto H, Yoshida H, Elia AJ, Samper E, Potter J, Wakeham A, Marengere L, Langille BL, Crabtree GR, Mak TW. Role of the NF-ATc transcription factor in morphogenesis of cardiac valves and septum. Nature. 1998;392:182-6.
    72. Guo L, Nakamura K, Lynch J, Opas M, Olson EN, Agellon LB, Michalak M. Cardiac-specific expression of calcineurin reverses embryonic lethality in calreticulin-deficient mouse. J Biol Chem. 2002;277:50776-9.
    73. Sola C, Tusell JM, Serratosa J. Comparative study of the distribution of calmodulin kinase II and calcineurin in the mouse brain. J Neurosci Res. 1999;57:651-62.
    74. Lim HW, Molkentin JD. Calcineurin and human heart failure. Nat Med. 1999;5:246-7.
    75. Ritter O, Hack S, Schuh K, Rothlein N, Perrot A, Osterziel KJ, Schulte HD, Neyses L. Calcineurin in human heart hypertrophy. Circulation. 2002; 105:2265-9.
    76. Morioka M, Nagahiro S, Fukunaga K, Miyamoto E, Ushio Y. Calcineurin in the adult rat hippocampus: different distribution in CA1 and CA3 subfields. Neuroscience. 1997;78:673-84.
    77. Usuda N, Arai H, Sasaki H, Hanai T, Nagata T, Muramatsu T, Kincaid RL, Higuchi S. Differential subcellular localization of neural isoforms of the catalytic subunit of calmodulin-dependent protein phosphatase (calcineurin) in central nervous system neurons: immunohistochemistry on formalin-fixed paraffin sections employing antigen retrieval by microwave irradiation. J Histochem Cytochem. 1996;44:13-8.
    78. Molkentin JD, Lu JR, Antos CL, Markham B, Richardson J, Robbins J, Grant SR, Olson EN. A calcineurin-dependent transcriptional pathway for cardiac hypertrophy. Cell. 1998;93:215-28.
    79. Lim HW, De Windt LJ, Steinberg L, Taigen T, Witt SA, Kimball TR, Molkentin JD. Calcineurin expression, activation, and function in cardiac pressure-overload hypertrophy. Circulation. 2000; 101:2431-7.
    80. Sussman MA, Lim HW, Gude N, Taigen T, Olson EN, Robbins J, Colbert MC, Gualberto A, Wieczorek DF, Molkentin JD. Prevention of cardiac hypertrophy in mice by calcineurin inhibition. Science. 1998;281:1690-3.
    81. Shimoyama M, Hayashi D, Takimoto E, Zou Y, Oka T, Uozumi H, Kudoh S, Shibasaki F, Yazaki Y, Nagai R, Komuro I. Calcineurin plays a critical role in pressure overload-induced cardiac hypertrophy. Circulation. 1999; 100:2449-54.
    82. Eto Y, Yonekura K, Sonoda M, Arai N, Sata M, Sugiura S, Takenaka K, Gualberto A, Hixon ML, Wagner MW, Aoyagi T. Calcineurin is activated in rat hearts with physiological left ventricular hypertrophy induced by voluntary exercise training. Circulation. 2000; 101:2134-7.
    83. Hayashida W, Kihara Y, Yasaka A, Sasayama S. Cardiac calcineurin during transition from hypertrophy to heart failure in rats. Biochem Biophys Res Commun. 2000;273:347-51.
    84. Meguro T, Hong C, Asai K, Takagi G, McKinsey TA, Olson EN, Vatner SF. Cyclosporine attenuates pressure-overload hypertrophy in mice while enhancing susceptibility to decompensation and heart failure. Circ Res. 1999;84:735-40.
    85. Zhang W, Kowal RC, Rusnak F, Sikkink RA, Olson EN, Victor RG. Failure of calcineurin inhibitors to prevent pressure-overload left ventricular hypertrophy in rats. Circ Res. 1999;84:722-8.
    86. Ding B, Price RL, Borg TK, Weinberg EO, Halloran PF, Lorell BH. Pressure overload induces severe hypertrophy in mice treated with cyclosporine, an inhibitor of calcineurin. Circ Res. 1999;84:729-34.
    87. Sussman MA, Welch S, Gude N, Khoury PR, Daniels SR, Kirkpatrick D, Walsh RA, Price RL, Lim HW, Molkentin JD. Pathogenesis of dilated cardiomyopathy: molecular, structural, and population analyses in tropomodulin-overexpressing transgenic mice. Am J Pathol. 1999;155:2101-13.
    88. Zhang W, Kowal RC, Rusnak F, Sikkink RA, Olson EN, Victor RG. Failure of calcineurin inhibitors to prevent pressure-overload left ventricular hypertrophy in rats. In: Circ Res; 1999:722-8.
    89. Fatkin D, McConnell BK, Mudd JO, Semsarian C, Moskowitz IG, Schoen FJ, Giewat M, Seidman CE, Seidman JG. An abnormal Ca(2+) response in mutant sarcomere protein-mediated familial hypertrophic cardiomyopathy. J Clin Invest. 2000; 106:1351-9.
    90. Li J, Yatani A, Kim SJ, Takagi G, Irie K, Zhang Q, Karoor V, Hong C, Yang G, Sadoshima J, Depre C, Vatner DE, West MJ, Vatner SF. Neurally-mediated increase in calcineurin activity regulates cardiac contractile function in absence of hypertrophy. Cardiovasc Res. 2003;59:649-57.
    91. Galindo-Fraga A, Arrieta O, Castillo-Martinez L, Narvaez R, Oseguera-Moguel J, Orea-Tejeda A. Elevation of plasmatic endothelin in patients with heart failure. Arch Med Res. 2003;34:367-72.
    92. Kjaer A, Appel J, Hildebrandt P, Petersen CL. Basal and exercise-induced neuroendocrine activation in patients with heart failure and in normal subjects. Eur J Heart Fail. 2004;6:29-39.
    93. Aronson D, Burger AJ. Neurohumoral activation and ventricular arrhythmias in patients with decompensated congestive heart failure: role of endothelin. Pacing Clin Electrophysiol. 2003;26:703-10.
    94. Zhu W, Zou Y, Shiojima I, Kudoh S, Aikawa R, Hayashi D, Mizukami M, Toko H, Shibasaki F, Yazaki Y, Nagai R, Komuro I. Ca2+/calmodulin-dependent kinase II and calcineurin play critical roles in endothelin-1-induced cardiomyocyte hypertrophy. J Biol Chem. 2000;275:15239-45.
    95. Clapham DE. Calcium signaling. Cell. 1995;80:259-68.
    96. Schmidt U, Hajjar RJ, Helm PA, Kim CS, Doye AA, Gwathmey JK. Contribution of abnormal sarcoplasmic reticulum ATPase activity to systolic and diastolic dysfunction in human heart failure. J Mol Cell Cardiol. 1998;30:1929-37.
    97. Balke CW, Shorofsky SR. Alterations in calcium handling in cardiac hypertrophy and heart failure. In: Cardiovasc Res; 1998:290-9.
    98. Crabtree GR. Generic signals and specific outcomes: signaling through Ca2+, calcineurin, and NF-AT. Cell. 1999;96:611-4.
    99. Balke CW, Shorofsky SR. Alterations in calcium handling in cardiac hypertrophy and heart failure. Cardiovasc Res. 1998;37:290-9.
    100. Yang SA, Klee CB. Low affinity Ca2+-binding sites of calcineurin B mediate conformational changes in calcineurin A. Biochemistry. 2000;39:16147-54.
    101. Stemmer PM, Klee CB. Dual calcium ion regulation of calcineurin by calmodulin and calcineurin B. In: Biochemistry; 1994:6859-66.
    102. Wang KK, Roufogalis BD, Villalobo A. Characterization of the fragmented forms of calcineurin produced by calpain I. Biochem Cell Biol. 1989;67:703-11.
    103. Tallant EA, Brumley LM, Wallace RW. Activation of a calmodulin-dependent phosphatase by a Ca2+-dependent protease. Biochemistry. 1988;27:2205-11.
    104. Manalan AS, Klee CB. Activation of calcineurin by limited proteolysis. Proc Natl Acad Sci USA. 1983;80:4291-5.
    105. Hubbard MJ, Klee CB. Characterization of a high-affinity monoclonal antibody to calcineurin whose epitope defines a new structural domain of calcineurin A. Eur J Biochem. 1989; 185:411-8.
    106. Yang SA, Klee C. Study of calcineurin structure by limited proteolysis. Methods Mol Biol. 2002;172:317-34.
    107. Thompson VF, Goll DE. Purification of mu-calpain, m-calpain, and calpastatin from animal tissues. Methods Mol Biol. 2000;144:3-16.
    108. Iizuka K, Kawaguchi H, Yasuda H. Calpain is activated during hypoxic myocardial cell injury. Biochem Med Metab Biol. 1991;46:427-31.
    109. Siman R, Noszek JC. Excitatory amino acids activate calpain I and induce structural protein breakdown in vivo. Neuron. 1988;1:279-87.
    110. Lee KS, Frank S, Vanderklish P, Arai A, Lynch G. Inhibition of proteolysis protects hippocampal neurons from ischemia. Proc Natl Acad Sci U S A. 1991;88:7233-7.
    111. Azuma M, David LL, Shearer TR. Cysteine protease inhibitor E64 reduces the rate of formation of selenite cataract in the whole animal. Curr Eye Res. 1991;10:657-66.
    112. Wang KK, Yuen PW. Calpain inhibition: an overview of its therapeutic potential. Trends Pharmacol Sci. 1994; 15:412-9.
    113. Aoki K, Imajoh S, Ohno S, Emori Y, Koike M, Kosaki G, Suzuki K. Complete amino acid sequence of the large subunit of the low-Ca2+-requiring form of human Ca2+-activated neutral protease (muCANP) deduced from its cDNA sequence. FEBS Lett. 1986;205:313-7.
    114. Sorimachi H, Ishiura S, Suzuki K. Structure and physiological function of calpains. Biochem J. 1997;328 ( Pt 3):721-32.
    115. Imajoh S, Kawasaki H, Suzuki K. Limited autolysis of calcium-activated neutral protease (CANP): reduction of the Ca2+-requirement is due to the NH2-terminal processing of the large subunit. J Biochem (Tokyo). 1986; 100:633-42.
    116. Saido TC, Shibata M, Takenawa T, Murofushi H, Suzuki K. Positive regulation of mu-calpain action by polyphosphoinositides. J Biol Chem. 1992;267:24585-90.
    117. Perrino BA, Ng LY, Soderling TR. Calcium regulation of calcineurin phosphatase activity by its B subunit and calmodulin. Role of the autoinhibitory domain. J Biol Chem. 1995;270:340-6.
    118. Klee CB, Ren H, Wang X. Regulation of the calmodulin-stimulated protein phosphatase, calcineurin. J Biol Chem. 1998;273:13367-70.
    119. Wu HY, Tomizawa K, Oda Y, Wei FY, Lu YF, Matsushita M, Li ST, Moriwaki A, Matsui H. Critical role of calpain-mediated cleavage of calcineurin in excitotoxic neurodegeneration. In: J Biol Chem; 2004:4929-40.
    120. Pollack JR, Witt RC, Sugimoto JT. Differential effects of calpain inhibitors on hypertrophy of cardiomyocytes. In: Mol Cell Biochem; 2003:47-50.
    121. Arthur GD, Belcastro AN. A calcium stimulated cysteine protease involved in isoproterenol induced cardiac hypertrophy. In: Mol Cell Biochem; 1997:241-8.
    122. Kim MJ, Jo DG, Hong GS, Kim BJ, Lai M, Cho DH, Kim KW, Bandyopadhyay A, Hong YM, Kim do H, Cho C, Liu JO, Snyder SH, Jung YK. Calpain-dependent cleavage of cain/cabin1 activates calcineurin to mediate calcium-triggered cell death. Proc Natl Acad Sci U S A. 2002;99:9870-5.
    123. Diedrichs H, Chi M, Boelck B, Mehlhorm U, Schwinger RH. Increased regulatory activity of the calcineurin/NFAT pathway in human heart failure. Eur J Heart Fail. 2004;6:3-9.
    124. Aitken A, Klee CB, Cohen P. The structure of the B subunit of calcineurin. Eur J Biochem. 1984; 139:663-71.
    125. Feng B, Stemmer PM. Interactions of calcineurin A, calcineurin B, and Ca2+. Biochemistry. 1999;38:12481-9.
    126. Wu H, Naya FJ, McKinsey TA, Mercer B, Shelton JM, Chin ER, Simard AR, Michel RN, Bassel-Duby R, Olson EN, Williams RS. MEF2 responds to multiple calcium-regulated signals in the control of skeletal muscle fiber type. EmboJ. 2000;19:1963-73.
    127. Verdin E, Dequiedt F, Kasler HG. Class II histone deacetylases: versatile regulators. Trends Genet. 2003;19:286-93.
    128. Rothermel BA, Vega RB, Williams RS. The role of modulatory calcineurin-interacting proteins in calcineurin signaling. Trends Cardiovasc Med.2003;13:15-21.
    129. Akhter SA, Luttrell LM, Rockman HA, Iaccarino G, Lefkowitz RJ, Koch WJ. Targeting the receptor-Gq interface to inhibit in vivo pressure overload myocardial hypertrophy. Science. 1998;280:574-7.
    130. D'Angelo DD, Sakata Y, Lorenz JN, Boivin GP, Walsh RA, Liggett SB, Dorn GW, 2nd. Transgenic Galphaq overexpression induces cardiac contractile failure in mice. Proc Natl Acad Sci U S A. 1997;94:8121-6.
    131. King MM, Huang CY. The calmodulin-dependent activation and deactivation of the phosphoprotein phosphatase, calcineurin, and the effect of nucleotides, pyrophosphate, and divalent metal ions. Identification of calcineurin as a Zn and Fe metalloenzyme. J Biol Chem. 1984;259:8847-56.
    132. Tsao L, Neville C, Musaro A, McCullagh KJ, Rosenthal N. Revisiting calcineurin and human heart failure. Nat Med. 2000;6:2-3.
    133. Lowry OH, Rosebrough NJ, Farr AL, Randall RJ. Protein measurement with the Folin phenol reagent. J Biol Chem. 1951;193:265-75.
    134. Dawson TM, Steiner JP, Lyons WE, Fotuhi M, Blue M, Snyder SH. The immunophilins, FK506 binding protein and cyclophilin, are discretely localized in the brain: relationship to calcineurin. Neuroscience. 1994;62:569-80.
    135. Polli JW, Billingsley ML, Kincaid RL. Expression of the calmodulin-dependent protein phosphatase, calcineurin, in rat brain: developmental patterns and the role of nigrostriatal innervation. Brain Res Dev Brain Res. 1991;63:105-19.
    136. Takahashi M, Tomizawa K, Ishiguro K. Distribution of tau protein kinase I/glycogen synthase kinase-3beta, phosphatases 2A and 2B, and phosphorylated tau in the developing rat brain. Brain Res. 2000;857:193-206.
    137. Nakamura A, Yoshida K, Takeda S, Dohi N, Ikeda S. Progression of dystrophic features and activation of mitogen-activated protein kinases and calcineurin by physical exercise, in hearts of mdx mice. FEBS Lett. 2002;520:18-24.
    138. Lakshmikuttyamma A, Selvakumar P, Kakkar R, Kanthan R, Wang R, Sharma RK. Activation of calcineurin expression in ischemia-reperfused rat heart and in human ischemic myocardium. J Cell Biochem. 2003;90:987-97.
    139. Haq S, Choukroun G, Lim H, Tymitz KM, del Monte F, Gwathmey J, Grazette L, Michael A, Hajjar R, Force T, Molkentin JD. Differential activation of signal transduction pathways in human hearts with hypertrophy versus advanced heart failure. Circulation. 2001;103:670-7.
    140. Hubbard MJ, Klee CB. Functional domain structure of calcineurin A: mapping by limited proteolysis. Biochemistry. 1989;28:1868-74.
    141. Yamazaki T, Komuro I, Kudoh S, Zou Y, Shiojima I, Hiroi Y, Mizuno T, Maemura K, Kurihara H, Aikawa R, Takano H, Yazaki Y. Endothelin-1 is involved in mechanical stress-induced cardiomyocyte hypertrophy. J Biol Chem. 1996;271:3221-8.
    142. Kamp TJ, Hell JW. Regulation of cardiac L-type calcium channels by protein kinase A and protein kinase C. Circ Res. 2000;87:1095-102.
    143. Puri TS, Gerhardstein BL, Zhao XL, Ladner MB, Hosey MM. Differential effects of subunit interactions on protein kinase A- and C-mediated phosphorylation of L-type calcium channels. Biochemistry. 1997;36:9605-15.
    144. Hashimoto Y, Soderling TR. Regulation of calcineurin by phosphorylation. Identification of the regulatory site phosphorylated by Ca2+/calmodulin-dependent protein kinase II and protein kinase C. J Biol Chem. 1989;264:16524-9.
    145. Tung HY. Phosphorylation of the calmodulin-dependent protein phosphatase by protein kinase C. Biochem Biophys Res Commun. 1986;138:783-8.
    146. Coghlan VM, Perrino BA, Howard M, Langeberg LK, Hicks JB, Gallatin WM, Scott JD. Association of protein kinase A and protein phosphatase 2B with a common anchoring protein. Science. 1995;267:108-11.
    147. Cohen P. The structure and regulation of protein phosphatases. Annu Rev Biochem. 1989;58:453-508.
    148. Hens JJ, De Wit M, Ghijsen WE, Leenders AG, Boddeke HW, Kissmehl R, Wiegant VM, Weller U, Gispen WH, De Graan PN. Role of calcineurin in Ca2+-induced release of catecholamines and neuropeptides. J Neurochem. 1998;71:1978-86.
    149. Shin HG, Barnett JV, Chang P, Reddy S, Drinkwater DC, Pierson RN, Wiley RG, Murray KT. Molecular heterogeneity of protein kinase C expression in human ventricle. Cardiovasc Res. 2000;48:285-99.
    150. Enz A, Shapiro G, Chappuis A, Dattler A. Nonradioactive assay for protein phosphatase 2B (calcineurin) activity using a partial sequence of the subunit of cAMP-dependent protein kinase as substrate. Anal Biochem. 1994;216:147-53.
    151. Donella-Deana A, Krinks MH, Ruzzene M, Klee C, Pinna LA. Dephosphorylation of phosphopeptides by calcineurin (protein phosphatase 2B). Eur J Biochem. 1994;219:109-17.
    152. Nishikawa K, Toker A, Johannes FJ, Songyang Z, Cantley LC. Determination of the specific substrate sequence motifs of protein kinase C isozymes. JBiol Chem. 1997;272:952-60.
    153. Herget T, Oehrlein SA, Pappin DJ, Rozengurt E, Parker PJ. The myristoylated alanine-rich C-kinase substrate (MARCKS) is sequentially phosphorylated by conventional, novel and atypical isotypes of protein kinase C. Eur J Biochem. 1995;233:448-57.
    154. Seki K, Chen HC, Huang KP. Dephosphorylation of protein kinase C substrates, neurogranin, neuromodulin, and MARCKS, by calcineurin and protein phosphatases 1 and 2A. Arch Biochem Biophys. 1995;316:673-9.
    155. Malbon CC, Tao J, Wang HY. AKAPs (A-kinase anchoring proteins) and molecules that compose their G-protein-coupled receptor signalling complexes. Biochem J. 2004;379:1-9.
    156. Sun L, Youn HD, Loh C, Stolow M, He W, Liu JO. Cabin 1, a negative regulator for calcineurin signaling in T lymphocytes. Immunity. 1998;8:703-11.
    157. Wu HY, Tomizawa K, Oda Y, Wei FY, Lu YF, Matsushita M, Li ST, Moriwaki A, Matsui H. Critical role of calpain-mediated cleavage of calcineurin in excitotoxic neurodegeneration. J Biol Chem. 2004;279:4929-40.
    158. Croall DE, DeMartino GN. Calcium-activated neutral protease (calpain) system: structure, function, and regulation. Physiol Rev. 1991;71:813-47.
    159. Croall DE, Moffett K, Hatch H. Casein zymography of calpains using a 4-(2-hydroxyethyl)-l-piperazineethanesulfonic acid-imidazole buffer. Anal Biochem. 2002;304:129-32.
    160. Rizo J, Sudhof TC. C2-domains, structure and function of a universal
     Ca2+-binding domain. J Biol Chem. 1998; 273: 15879-82.
    161. Khorchid A, Ikura M. How calpain is activated by calcium. In: Nat Struct Biol; 2002: 239-41.
    162. Khorchid A, Ikura M. How calpain is activated by calcium. Nat Struct Biol. 2002; 9: 239-41.
    163. Suzuki K, Tsuji S, Kubota S, Kimura Y, Imahori K. Limited autolysis of Ca2+-activated neutral protease (CANP) changes its sensitivity to Ca2+ ions. J Biochem (Tokyo). 1981; 90: 275-8.
    164. Suzuki K, Tsuji S, Ishiura S, Kimura Y, Kubota S, Imahori K. Autolysis of calcium-activated neutral protease of chicken skeletal muscle. J Biochem (Tokyo). 1981; 90: 1787-93.
    165. Dayton WR. Comparison of low-and high-calcium-requiring forms of the calcium-activated protease with their autocatalytic breakdown products. Biochim Biophys Acta. 1982; 709: 166-72.
    166. Thompson VF, Lawson K, Goll DE. Effect of mu-calpain on m-calpain. Biochem Biophys Res Commun. 2000; 267: 495-9.
    167. De Windt LJ, Lim HW, Bueno OF, Liang Q, Delling U, Braz JC, Glascock BJ, Kimball TF, del Monte F, Hajjar RJ, Molkentin JD. Targeted inhibition of calcineurin attenuates cardiac hypertrophy in vivo. Proc Natl Acad Sci U S A. 2001; 98: 3322-7.
    168. Kishimoto A, Kajikawa N, Shiota M, Nishizuka Y. Proteolytic activation of calcium-activated, phospholipid-dependent protein kinase by calcium-dependent neutral protease. J Biol Chem. 1983; 258: 1156-64.
    169. Luo JH, Kahn S, O'Driscoll K, Weinstein IB. The regulatory domain of protein kinase C beta 1 contains phosphatidylserine-and phorbol ester-dependent calcium binding activity. J Biol Chem. 1993; 268: 3715-9.
    170. Saido TC, Sorimachi H, Suzuki K. Calpain: new perspectives in molecular diversity and physiological-pathological involvement. Faseb J. 1994; 8: 814-22.
    171. Nishizuka Y. Studies and perspectives of protein kinase C. Science. 1986;233:305-12.
    172. Sakon M, Kambayashi J, Ohno H, Kosaki G. Two forms of Ca++-activated neutral protease in platelets. Thromb Res. 1981;24:207-14.
    173. Kieran D, Greensmith L. Inhibition of calpains, by treatment with leupeptin, improves motoneuron survival and muscle function in models of motoneuron degeneration. Neuroscience. 2004; 125:427-39.
    174. Arthur GD, Belcastro AN. A calcium stimulated cysteine protease involved in isoproterenol induced cardiac hypertrophy. Mol Cell Biochem. 1997;176:241-8.
    175. Pollack JR, Witt RC, Sugimoto JT. Differential effects of calpain inhibitors on hypertrophy of cardiomyocytes. Mol Cell Biochem. 2003;251:47-50.
    176. Melloni E, Minafra R, Salamino F, Pontremoli S. Properties and intracellular localization of calpain activator protein. Biochem Biophys Res Commun. 2000;272:472-6.
    177. Wang HG, Pathan N, Ethell IM, Krajewski S, Yamaguchi Y, Shibasaki F, McKeon F, Bobo T, Franke TF, Reed JC. Ca2+-induced apoptosis through calcineurin dephosphorylation of BAD. Science. 1999;284:339-43.
    178. Shibasaki F, McKeon F. Calcineurin functions in Ca(2+)-activated cell death in mammalian cells. J Cell Biol. 1995; 131:735-43.
    179. Pasquet JM, Dachary-Prigent J, Nurden AT. Calcium influx is a determining factor of calpain activation and microparticle formation in platelets. Eur J Biochem. 1996;239:647-54.
    180. Al Z, Cohen CM. Phorbol 12-myristate 13-acetate-stimulated phosphorylation of erythrocyte membrane skeletal proteins is blocked by calpain inhibitors: possible role of protein kinase M. Biochem J. 1993;296 (Pt3):675-83.
    181. Kissinger CR, Parge HE, Knighton DR, Lewis CT, Pelletier LA, Tempczyk A, Kalish VJ, Tucker KD, Showalter RE, Moomaw EW, et al. Crystal structures of human calcineurin and the human FKBP12-FK506-calcineurin complex. Nature. 1995;378:641-4.

© 2004-2018 中国地质图书馆版权所有 京ICP备05064691号 京公网安备11010802017129号

地址:北京市海淀区学院路29号 邮编:100083

电话:办公室:(+86 10)66554848;文献借阅、咨询服务、科技查新:66554700