Improving vagal activity ameliorates cardiac fibrosis induced by angiotensin II: in vivo and in vitro

Jin-Jun Liu, Ning Huang, Yi Lu, Mei Zhao, Xiao-Jiang Yu, Yang Yang, Yong-hua Yang, Wei-Jin Zang, Jin-Jun Liu, Ning Huang, Yi Lu, Mei Zhao, Xiao-Jiang Yu, Yang Yang, Yong-hua Yang, Wei-Jin Zang

Abstract

Cardiac remodeling is characterized by overactivity of the renin-angiotensin system (RAS) and withdrawal of vagal activity. We hypothesized that improving vagal activity could attenuate cardiac fibrosis induced by angiotensin II (Ang II) in vivo and in vitro. Rats were subjected to abdominal aorta constriction (AAC) with or without pyridostigmine (PYR) (31 mg/kg/d). After 8 weeks, PYR significantly decreased Ang II level, AT1 protein expression, and collagen deposition in cardiac tissue and improved heart rate variability, baroreflex sensitivity and cardiac function, which were abolished by atropine. In vitro, treatment of cardiac fibroblasts (CFs) with Ang II (10(-7) M) increased cell proliferation, migration, transformation, and secretory properties, which were significantly diminished by acetylcholine (ACh, 10(-6) M). Subsequently, Ang II significantly increased collagen type I expression as well as metalloproteinase (MMP)-2 expression and activity. Transforming growth factor (TGF)-β1 expression and Smad3 phosphorylation presented a similar trend. Notably, the knockdown of the acetylcholine M2 receptor by siRNA could abolish ACh anti-fibrotic action. These data implicated cholinesterase inhibitor can increase vagal activity and reduce local Ang II level, and ACh inhibit Ang II pro-fibrotic effects. Our findings suggested that the parasympathetic nervous system can serve as a promising target for cardiac remodeling treatment.

Figures

Figure 1. PYR inhibits cardiac fibrosis and…
Figure 1. PYR inhibits cardiac fibrosis and improves cardiac function.
(a) The image shows Masson’s trichrome stained collagen in the cardiac interstitium. Blue parts represent collagen. Bar = 50 μm. (b) Representative LV sections stained with picrosirius red for collagen type I (red) and III (orange yellow) from all groups as indicated. Bar = 25 μm. (c,d) LV tissues were analyzed for collagen type I and III by western blot. (e) MAP, mean arterial pressure. (e) HR, heart rate. (f) LVSP, left ventricular systolic pressure. (h) LVEDP, left ventricular end-diastolic pressure. Data are presented as means ± SEM. *P < 0.05, **P < 0.01 vs. control group; #P < 0.05 vs. AAC group; &P < 0.05 vs. PYR group.
Figure 2. PYR decreases Ang II level…
Figure 2. PYR decreases Ang II level and improves vagal activity.
(a) Radioimmunoassay was performed to detect Ang II in serum and (b) left ventricular tissues. (c) Western blot and quantitative evaluation of AT1 expression. (d) Baroreflex sensitivity (BRS). (e) Standard deviation of RR Interval (SDNN). (f) Normalized low frequency power (LF power; %). (g) Normalized high frequency power (HF power, %). (h) Absolute values of low/high frequency power ratio (LF/HF). Data are presented as means ± SEM. **P < 0.01 and ***P < 0.001 vs. control group; #P < 0.05 and # #P < 0.01 vs. AAC group; &P < 0.05 and &&P < 0.01 vs. PYR group.
Figure 3. ACh attenuates Ang II-induced collagen…
Figure 3. ACh attenuates Ang II-induced collagen production.
(a) Fluorescence micrographs of vimentin, Von Willebrand Factor and desmin. Bar = 25 μm. (b,c) For collagen type I assay, CFs were cultured in the indicated experimental conditions and immunostained with an antibody against collagen I (green), and the histograms showed the corresponding densitometric analyses of the intensity of collagen I fluorescence signal. Bar = 50 μm. (d) Western blot analysis of collagen I expression in CFs. The densitometric analysis of the bands normalized to GAPDH is reported in the histograms. (e) The content of hydroxyproline was measured by using a bioluminescent assay. (f) MMP-2 activity was measured by gelatin zymography. (g) Western blot and quantitative evaluation of MMP-2 expression. Data are presented as means ± SEM. **P < 0.01 vs. control group; #P < 0.05 and # #P < 0.01 vs. Ang II group; &P < 0.05 and &&P < 0.01vs. ACh group.
Figure 4. ACh suppresses Ang II-induced proliferation…
Figure 4. ACh suppresses Ang II-induced proliferation and viability.
(a) Representative photomicrographs of 5-ethynil-2-deoxyuridine (EdU) staining (left panel) and corresponding total cell photomicrographs (middle panel). Proliferative cell nuclei were labeled with EdU (green). Cell nuclei were labeled by using PI (Red). (b) Effect of ACh on cell proliferation as determined by EdU staining. Quantitative data showing the percentage of EdU-positive cells (number of green vs. red nuclei). Bar = 50 μm. Data are presented as means ± SEM. *P < 0.05 and **P < 0.01 vs. control group; ##P < 0.01 vs. Ang II group; &P < 0.05 vs. ACh group.
Figure 5. ACh inhibits CFs migration and…
Figure 5. ACh inhibits CFs migration and transformation induced by Ang II.
(a,b) Fluorescence immunohistochemistry using a specific α-SMA first antibody following by a second antibody conjugated to FITC was performed to demonstrate fibroblast transformation induced by Ang II. Nuclei were stained with PI. Bar = 25 μm. (c,d) Representative pictures of cell migration detected by transwell migration assay. Bar = 50 μm. Data are presented as means ± SEM. **P < 0.01 and ***P < 0.001 vs. control; ##P < 0.001 vs. Ang II; &P < 0.05 and &&P < 0.01 vs. ACh.
Figure 6. ACh inhibits the Ang II-induced…
Figure 6. ACh inhibits the Ang II-induced activation of the TGF-β1/Smad3 pathway.
(a) Immunofluroescence images show that ACh blocks Smad3 nuclear translocation induced by Ang II. (b,c) Western blot evaluated expression of TGF-β1 and P-Smad3. Bar = 25 μm. Data are presented as means ± SEM. *P < 0.05 and **P < 0.01 vs. control; #P < 0.05 and ##P < 0.01 vs. Ang II; &P < 0.05 and &&P < 0.01 vs. ACh.
Figure 7. M 2 AChR knockdown abrogates…
Figure 7. M2 AChR knockdown abrogates ACh anti-fibrotic effect.
(a) Western blot was performed to assess M2 AChR knockdown. (b) Hydroxyproline content was measured by using a bioluminescent assay. (c) M2 AChR siRNA transfection increases collagen I contents in the presence of ACh in Ang II treated cells. Bar = 50 μm. (d) M2 AChR siRNA transfection increases fibroblasts migration. Bar = 50 μm. (e) Cell proliferation was measured as the percentage of EdU-positive cells. Bar = 50 μm. (f) M2 AChR siRNA transfection increases fibroblast transformation. Bar = 25 μm. (g) Expression of TGF-β1, P-Smad3, and collagen I. Data are presented as means ± SEM. *P < 0.05 and **P < 0.01 vs. control siRNA group.

References

    1. Mehta P. K. & Griendling K. K. Angiotensin II cell signaling: physiological and pathological effects in the cardiovascular system. Am J Physiol Cell Physiol 292, C82 (2007).
    1. Kumar R., Singh V. P. & Baker K. M. The intracellular renin-angiotensin system: implications in cardiovascular remodeling. Curr Opin Nephrol Hypertens 17, 168 (2008).
    1. Yamamoto K. et al. Deletion of angiotensin-converting enzyme 2 accelerates pressure overload-induced cardiac dysfunction by increasing local angiotensin II. HYPERTENSION 47, 718 (2006).
    1. Villarreal F. J., Kim N. N., Ungab G. D., Printz M. P. & Dillmann W. H. Identification of functional angiotensin II receptors on rat cardiac fibroblasts. CIRCULATION 88, 2849 (1993).
    1. Nakano A. & Kitakaze M. [Use of angiotensin type II receptor blockers as a potential strategy for chronic heart failure]. Nihon Rinsho 64, 910 (2006).
    1. Makowski K. et al. Autonomic nervous system and left ventricular hypertrophy in essential hypertension. KARDIOL POL 57, 520, 532 (2002).
    1. Thayer J. F. & Lane R. D. The role of vagal function in the risk for cardiovascular disease and mortality. BIOL PSYCHOL 74, 224 (2007).
    1. Kishi T. Heart failure as an autonomic nervous system dysfunction. J CARDIOL 59, 117 (2012).
    1. Vanoli E. et al. Vagal stimulation and prevention of sudden death in conscious dogs with a healed myocardial infarction. CIRC RES 68, 1471 (1991).
    1. Li M. et al. Vagal nerve stimulation markedly improves long-term survival after chronic heart failure in rats. CIRCULATION 109, 120 (2004).
    1. Handa T. et al. Anti-Alzheimer’s drug, donepezil, markedly improves long-term survival after chronic heart failure in mice. J CARD FAIL 15, 805 (2009).
    1. Sabino J. P., Da S. C., de Melo R. F., Fazan R. J. & Salgado H. C. The treatment with pyridostigmine improves the cardiocirculatory function in rats with chronic heart failure. Auton Neurosci 173, 58 (2013).
    1. Porter K. E. & Turner N. A. Cardiac fibroblasts: at the heart of myocardial remodeling. Pharmacol Ther 123, 255 (2009).
    1. Fan D., Takawale A., Lee J. & Kassiri Z. Cardiac fibroblasts, fibrosis and extracellular matrix remodeling in heart disease. Fibrogenesis Tissue Repair 5, 15 (2012).
    1. Peng J., Gurantz D., Tran V., Cowling R. T. & Greenberg B. H. Tumor necrosis factor-alpha-induced AT1 receptor upregulation enhances angiotensin II-mediated cardiac fibroblast responses that favor fibrosis. CIRC RES 91, 1119 (2002).
    1. Rocha-Resende C. et al. Non-neuronal cholinergic machinery present in cardiomyocytes offsets hypertrophic signals. J MOL CELL CARDIOL 53, 206 (2012).
    1. Matthiesen S. et al. Muscarinic receptors mediate stimulation of human lung fibroblast proliferation. Am J Respir Cell Mol Biol 35, 621 (2006).
    1. Liu J. J. et al. Acetylcholine prevents angiotensin II-induced oxidative stress and apoptosis in H9c2 cells. APOPTOSIS 16, 94 (2011).
    1. Werner C. M. & Bohm M. The therapeutic role of RAS blockade in chronic heart failure. Ther Adv Cardiovasc Dis 2, 167 (2008).
    1. Sun Y., Zhang J. Q., Zhang J. & Ramires F. J. Angiotensin II, transforming growth factor-beta1 and repair in the infarcted heart. J MOL CELL CARDIOL 30, 1559 (1998).
    1. Mortara A. et al. Arterial baroreflex modulation of heart rate in chronic heart failure: clinical and hemodynamic correlates and prognostic implications. CIRCULATION 96, 3450 (1997).
    1. Adamson P. B. et al. Continuous autonomic assessment in patients with symptomatic heart failure: prognostic value of heart rate variability measured by an implanted cardiac resynchronization device. CIRCULATION 110, 2389 (2004).
    1. Lu Y. et al. Pyridostigmine ameliorates cardiac remodeling induced by myocardial infarction via inhibition of the transforming growth factor-beta1/TGF-beta1-activated kinase pathway. J Cardiovasc Pharmacol 63, 412 (2014).
    1. Lataro R. M. et al. Increase in parasympathetic tone by pyridostigmine prevents ventricular dysfunction during the onset of heart failure. American journal of physiology. Regulatory, integrative and comparative physiology 305, R908–916 (2013).
    1. De La Fuente R. N. et al. Cholinergic stimulation with pyridostigmine improves autonomic function in infarcted rats. Clinical and experimental pharmacology & physiology 40, 610–616 (2013).
    1. Gavioli M. et al. Cholinergic signaling exerts protective effects in models of sympathetic hyperactivity-induced cardiac dysfunction. PloS one 9, e100179 (2014).
    1. Deb A. & Ubil E. Cardiac fibroblast in development and wound healing. J MOL CELL CARDIOL 70, 47 (2014).
    1. Van den Borne S. W. et al. Myocardial remodeling after infarction: the role of myofibroblasts. NAT REV CARDIOL 7, 30 (2010).
    1. Kim M. H., Kim M. O., Heo J. S., Kim J. S. & Han H. J. Acetylcholine inhibits long-term hypoxia-induced apoptosis by suppressing the oxidative stress-mediated MAPKs activation as well as regulation of Bcl-2, c-IAPs, and caspase-3 in mouse embryonic stem cells. APOPTOSIS 13, 295 (2008).
    1. Li D. L. et al. Acetylcholine inhibits hypoxia-induced tumor necrosis factor-alpha production via regulation of MAPKs phosphorylation in cardiomyocytes. J CELL PHYSIOL 226, 1052 (2011).
    1. Ma Y. et al. Myofibroblasts and the extracellular matrix network in post-myocardial infarction cardiac remodeling. Pflugers Arch 466, 1113 (2014).
    1. Weber K. T., Sun Y., Bhattacharya S. K., Ahokas R. A. & Gerling I. C. Myofibroblast-mediated mechanisms of pathological remodelling of the heart. NAT REV CARDIOL 10, 15 (2013).
    1. Siddesha J. M. et al. Angiotensin II stimulates cardiac fibroblast migration via the differential regulation of matrixins and RECK. J MOL CELL CARDIOL 65, 9 (2013).
    1. Ruiz-Ortega M., Ruperez M., Esteban V. & Egido J. Molecular mechanisms of angiotensin II-induced vascular injury. CURR HYPERTENS REP 5, 73 (2003).
    1. Campbell S. E. & Katwa L. C. Angiotensin II stimulated expression of transforming growth factor-beta1 in cardiac fibroblasts and myofibroblasts. J MOL CELL CARDIOL 29, 1947 (1997).
    1. Li L. et al. Angiotensin II increases periostin expression via Ras/p38 MAPK/CREB and ERK1/2/TGF-beta1 pathways in cardiac fibroblasts. CARDIOVASC RES 91, 80 (2011).
    1. Border W. A. & Noble N. A. Interactions of transforming growth factor-beta and angiotensin II in renal fibrosis. HYPERTENSION 31, 181 (1998).
    1. Schmierer B. & Hill C. S. TGFbeta-SMAD signal transduction: molecular specificity and functional flexibility. Nat Rev Mol Cell Biol 8, 970 (2007).
    1. Bujak M. et al. Essential role of Smad3 in infarct healing and in the pathogenesis of cardiac remodeling. CIRCULATION 116, 2127 (2007).
    1. Cooke J. P. Angiogenesis and the role of the endothelial nicotinic acetylcholine receptor. LIFE SCI 80, 2347 (2007).
    1. Yang B. et al. Choline produces cytoprotective effects against ischemic myocardial injuries: evidence for the role of cardiac m3 subtype muscarinic acetylcholine receptors. CELL PHYSIOL BIOCHEM 16, 163 (2005).
    1. Chen B. et al. MG132, a proteasome inhibitor, attenuates pressure-overload-induced cardiac hypertrophy in rats by modulation of mitogen-activated protein kinase signals. Acta Biochim Biophys Sin (Shanghai) 42, 253 (2010).
    1. Parthasarathy A. et al. Suppression of atrial natriuretic peptide/natriuretic peptide receptor-A-mediated signaling upregulates angiotensin-II-induced collagen synthesis in adult cardiac fibroblasts. MOL CELL BIOCHEM 378, 217 (2013).
    1. Yu J. G. et al. Baroreflex deficiency hampers angiogenesis after myocardial infarction via acetylcholine-alpha7-nicotinic ACh receptor in rats. EUR HEART J 34, 2412 (2013).
    1. Li D. et al. LOX-1 mediates oxidized low-density lipoprotein-induced expression of matrix metalloproteinases in human coronary artery endothelial cells. CIRCULATION 107, 612 (2003).
    1. Valente A. J. et al. Interleukin-17A stimulates cardiac fibroblast proliferation and migration via negative regulation of the dual-specificity phosphatase MKP-1/DUSP-1. CELL SIGNAL 24, 560 (2012).

Source: PubMed

3
Abonner