51
|
Zhang S, Bian H, Li X, Wu H, Bi Q, Yan Y, Wang Y. Hydrogen sulfide promotes cell proliferation of oral cancer through activation of the COX2/AKT/ERK1/2 axis. Oncol Rep 2016; 35:2825-32. [PMID: 26987083 DOI: 10.3892/or.2016.4691] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2015] [Accepted: 01/27/2016] [Indexed: 11/05/2022] Open
Abstract
Hydrogen sulfide, the third gaseous transmitter, is one of the main causes of halitosis in the oral cavity. It is generally considered as playing a deleterious role in many oral diseases including oral cancer. However, the regulatory mechanisms involved in the effects of hydrogen sulfide on oral cancer growth remain largely unknown. In the present study, we investigated the underlying mechanisms through CCK-8 assay, EdU incorporation, real-time PCR, western blot and pathway blockade assays. Our results showed that hydrogen sulfide promoted oral cancer cell proliferation through activation of the COX2, AKT and ERK1/2 pathways in a dose-dependent manner. Blocking any of the three above pathways inhibited hydrogen sulfide-induced oral cancer cell proliferation. Meanwhile, blockade of COX2 by niflumic acid downregulated NaHS-induced p-ERK and p-AKT expression. Inactivation of the AKT pathway by GSK690693 significantly decreased NaHS‑induced p-ERK1/2 expression, and inhibition of the ERK1/2 pathway by U0126 markedly increased NaHS-induced p-AKT expression. Either the AKT or ERK1/2 inhibitor did not significantly alter the COX2 expression level. Our data revealed, for the first time, that hydrogen sulfide promotes oral cancer cell proliferation through activation of the COX2/AKT/ERK1/2 axis, suggesting new potential targets to eliminate the effect of hydrogen sulfide on the development of oral cancer.
Collapse
Affiliation(s)
- Shuai Zhang
- Central Laboratory, Peking University School and Hospital of Stomatology, Beijing, P.R. China
| | - Huan Bian
- Department of Stomatology, The First Affiliated Hospital of the Chinese PLA General Hospital, Beijing, P.R. China
| | - Xiaoxu Li
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing, P.R. China
| | - Huanhuan Wu
- The Second Dental Center, Peking University School and Hospital of Stomatology, Beijing, P.R. China
| | - Qingwei Bi
- Department of Oral Surgery, Hospital for Oral Disease Prevention and Treatment, Harbin, Heilongjiang, P.R. China
| | - Yingbin Yan
- Department of Oral and Maxillofacial Surgery, Tianjin Stomatological Hospital, Tianjin, P.R. China
| | - Yixiang Wang
- Central Laboratory, Peking University School and Hospital of Stomatology, Beijing, P.R. China
| |
Collapse
|
52
|
Prevention of vascular smooth muscle cell proliferation and injury-induced neointimal hyperplasia by CREB-mediated p21 induction: An insight from a plant polyphenol. Biochem Pharmacol 2016; 103:40-52. [PMID: 26807478 DOI: 10.1016/j.bcp.2016.01.015] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Accepted: 01/13/2016] [Indexed: 12/23/2022]
Abstract
Cyclic adenosine monophosphate (cAMP)/protein kinase A (PKA)/cAMP response element (CRE)-binding protein (CREB) signaling cascade negatively regulates platelet-derived growth factor BB (PDGF-BB)-induced smooth muscle cell (SMC) proliferation, which is a critical event in the initiation and development of restenosis and atherosclerotic lesions. Salvianolic acid A (SAA) is one of the most abundant polyphenols extracted from salvia. The aim of this study is to investigate whether SAA exerts an action on PDGF-BB-induced proliferation via cAMP/PKA/CREB mechanism. SAA blunts PDGF-BB-induced human umbilical artery smooth muscle cell (hUASMC) proliferation via p21 induction, as evidenced by its increased mRNA and protein expression levels. The SAA-induced upregulation of p21 involves the cAMP/PKA signaling pathway; a cAMP analog mimicked the effects of SAA and a specific cAMP/PKA inhibitor opposed these effects. SAA also activated CREB, including phosphorylation at Ser133, and induced its nuclear translocation. Deletion and mutational analysis of p21 promoters, co-immunoprecipitation, and western blot analysis showed that CRE is essential for SAA-induced p21 protein expression. Transfection of dominant-negative CREB (mutated Ser133) plasmids into hUASMCs attenuated SAA-stimulated p21 expression. SAA upregulated p21 expression and activated CREB in the neointima of balloon-injured arteries in vivo. Our results indicate that SAA promotes p21 expression in SMCs through the cAMP/PKA/CREB signaling cascade in vitro and prevents injury-induced neointimal hyperplasia.
Collapse
|
53
|
Han JH, Kim Y, Jung SH, Lee JJ, Park HS, Song GY, Cuong NM, Kim YH, Myung CS. Murrayafoline A Induces a G0/G1-Phase Arrest in Platelet-Derived Growth Factor-Stimulated Vascular Smooth Muscle Cells. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2015; 19:421-6. [PMID: 26330754 PMCID: PMC4553401 DOI: 10.4196/kjpp.2015.19.5.421] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/19/2015] [Revised: 07/13/2015] [Accepted: 07/24/2015] [Indexed: 11/15/2022]
Abstract
The increased potential for vascular smooth muscle cell (VSMC) growth is a key abnormality in the development of atherosclerosis and post-angioplasty restenosis. Abnormally high activity of platelet-derived growth factor (PDGF) is believed to play a central role in the etiology of these pathophysiological situations. Here, we investigated the anti-proliferative effects and possible mechanism(s) of murrayafoline A, a carbazole alkaloid isolated from Glycosmis stenocarpa Guillamin (Rutaceae), on PDGF-BB-stimulated VSMCs. Murrayafoline A inhibited the PDGF-BB-stimulated proliferation of VSMCs in a concentration-dependent manner, as measured using a non-radioactive colorimetric WST-1 assay and direct cell counting. Furthermore, murrayafoline A suppressed the PDGF-BB-stimulated progression through G0/G1 to S phase of the cell cycle, as measured by [3H]-thymidine incorporation assay and cell cycle progression analysis. This anti-proliferative action of murrayafoline A, arresting cell cycle progression at G0/G1 phase in PDGF-BB-stimulated VSMCs, was mediated via down-regulation of the expression of cyclin D1, cyclin E, cyclin-dependent kinase (CDK)2, CDK4, and proliferating cell nuclear antigen (PCNA), and the phosphorylation of retinoblastoma protein (pRb). These results indicate that murrayafoline A may be useful in preventing the progression of vascular complications such as restenosis after percutaneous transluminal coronary angioplasty and atherosclerosis.
Collapse
Affiliation(s)
- Joo-Hui Han
- Department of Pharmacology, Chungnam National University College of Pharmacy, Daejeon 305-764, Korea
| | - Yohan Kim
- Department of Pharmacology, Chungnam National University College of Pharmacy, Daejeon 305-764, Korea
| | - Sang-Hyuk Jung
- Department of Pharmacology, Chungnam National University College of Pharmacy, Daejeon 305-764, Korea
| | - Jung-Jin Lee
- KM Application Center, Korea Institute of Oriental Medicine, Daegu 701-300, Korea
| | - Hyun-Soo Park
- Department of Pharmacology, Chungnam National University College of Pharmacy, Daejeon 305-764, Korea
| | - Gyu-Yong Song
- Department of Medicinal Chemistry, Chungnam National University College of Pharmacy, Daejeon 305-764, Korea. ; Institute of Drug Research & Development, Chungnam National University, Daejeon 305-764, Korea
| | - Nguyen Manh Cuong
- Institute of Natural Products Chemistry, Vietnam Academy of Science and Technology (VAST), 18 Hoang Quoc Viet St., 122100 Caugiay, Hanoi, Vietnam
| | - Young Ho Kim
- Institute of Drug Research & Development, Chungnam National University, Daejeon 305-764, Korea. ; Department of Natural Product Chemistry, Chungnam National University College of Pharmacy, Daejeon 305-764, Korea
| | - Chang-Seon Myung
- Department of Pharmacology, Chungnam National University College of Pharmacy, Daejeon 305-764, Korea. ; Institute of Drug Research & Development, Chungnam National University, Daejeon 305-764, Korea
| |
Collapse
|
54
|
Ricci C, Ferri N. Naturally occurring PDGF receptor inhibitors with potential anti-atherosclerotic properties. Vascul Pharmacol 2015; 70:1-7. [DOI: 10.1016/j.vph.2015.02.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Revised: 02/18/2015] [Accepted: 02/20/2015] [Indexed: 01/03/2023]
|
55
|
Li L, Zhou X, Li N, Sun M, Lv J, Xu Z. Herbal drugs against cardiovascular disease: traditional medicine and modern development. Drug Discov Today 2015; 20:1074-86. [PMID: 25956424 DOI: 10.1016/j.drudis.2015.04.009] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2015] [Revised: 03/23/2015] [Accepted: 04/28/2015] [Indexed: 12/31/2022]
Abstract
Herbal products have been used as conventional medicines for thousands of years, particularly in Eastern countries. Thousands of clinical and experimental investigations have focused on the effects and mechanisms-of-action of herbal medicine in the treatment of cardiovascular diseases (CVDs). Considering the history of clinical practice and the great potentials of herb medicine and/or its ingredients, a review on this topic would be helpful. This article discusses possible effects of herbal remedies in the prevention and treatment of CVDs. Crucially, we also summarize some underlying pharmacological mechanisms for herb products in cardiovascular regulations, which might provide interesting information for further understanding the effects of herbal medicines, and boost the prospect of new herbal products against CVDs.
Collapse
Affiliation(s)
- Lingjun Li
- Institute for Fetology, First Hospital of Soochow University, Suzhou 215006, China
| | - Xiuwen Zhou
- Institute for Fetology, First Hospital of Soochow University, Suzhou 215006, China
| | - Na Li
- Institute for Fetology, First Hospital of Soochow University, Suzhou 215006, China
| | - Miao Sun
- Institute for Fetology, First Hospital of Soochow University, Suzhou 215006, China
| | - Juanxiu Lv
- Institute for Fetology, First Hospital of Soochow University, Suzhou 215006, China
| | - Zhice Xu
- Institute for Fetology, First Hospital of Soochow University, Suzhou 215006, China; Center for Perinatal Biology, Division of Pharmacology, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA.
| |
Collapse
|
56
|
Luan Y, Chao S, Ju ZY, Wang J, Xue X, Qi TG, Cheng GH, Kong F. Therapeutic effects of baicalin on monocrotaline-induced pulmonary arterial hypertension by inhibiting inflammatory response. Int Immunopharmacol 2015; 26:188-93. [DOI: 10.1016/j.intimp.2015.01.009] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Revised: 01/04/2015] [Accepted: 01/09/2015] [Indexed: 11/30/2022]
|
57
|
Liu P, Yan S, Chen M, Chen A, Yao D, Xu X, Cai X, Wang L, Huang X. Effects of baicalin on collagen Ι and collagen ΙΙΙ expression in pulmonary arteries of rats with hypoxic pulmonary hypertension. Int J Mol Med 2015; 35:901-8. [PMID: 25716558 PMCID: PMC4356435 DOI: 10.3892/ijmm.2015.2110] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2014] [Accepted: 02/13/2015] [Indexed: 01/10/2023] Open
Abstract
The synthesis and accumulation of collagen play an important role in the formation and progression of hypoxic pulmonary hypertension. Baicalin has been reported to prevent bleomycin-induced pulmonary fibrosis. However, the role of baicalin in the treatment of pulmonary hypertension remains unknown. A disintegrin and metalloprotease with thrombospondin type-1 motif (ADAMTS-1) is a secreted enzyme that acts on a wide variety of extracellular matrix (ECM) substrates associated with vascular diseases. In this study, we aimed to investigate the effects of baicalin on the synthesis of collagen I in rats with pulmonary hypertension induced by hypoxia and the changes in ADAMTS-1 expression. A total of 24 Sprague Dawley rats were randomly assigned to 3 groups as follows: the control group (C), the hypoxia group (H) and the hypoxia + baicalin group (B). The rats in groups H and B were kept in a normobaric hypoxic chamber for 4 weeks, and the rats in group C were exposed to room air. We measured the hemodynamic indexes, including mean pulmonary artery pressure (mPAP), mean systemic (carotid) artery pressure (mSAP), and then calculated the mass ratio of right ventricle to left ventricle plus septum [RV/(LV + S)] to reflect the extent of right ventricular hypertrophy. We measured the mRNA and protein expression levels of type I collagen, type III collagen and ADAMTS-1 by hybridization in situ, and immunohistochemistry and western blot analysis, respectively. The results revealed that treatment with baicalin significantly reduced pulmonary artery pressure and attenuated the remodeling of the pulmonary artery under hypoxic conditions by increasing the expression of ADAMTS-1, so that the synthesis of type I collagen and its mRNA expression were inhibited. In conclusion, baicalin effectively inhibits the synthesis of collagen I in pulmonary arteries and this is associated with an increase in the expression of ADAMTS-1. Thus, treatment with baicalin may be an effective method for lowering pulmonary artery pressure and preventing pulmonary artery remodeling.
Collapse
Affiliation(s)
- Panpan Liu
- Intensive Care Unit, Ningbo Medical Treatment Center Lihuili Hospital, Ningbo, Zhejiang 315040, P.R. China
| | - Shuangquan Yan
- Division of Respiratory Medicine, Taizhou Enze Medical Center Luqiao Hospital, Taizhou, Zhejiang 318050, P.R. China
| | - Mayun Chen
- Division of Pulmonary Medicine, The First Affiliated Hospital of Wenzhou Medical University and Key Laboratory of Heart and Lung, Wenzhou, Zhejiang 325035, P.R. China
| | - Ali Chen
- Division of Pulmonary Medicine, The First Affiliated Hospital of Wenzhou Medical University and Key Laboratory of Heart and Lung, Wenzhou, Zhejiang 325035, P.R. China
| | - Dan Yao
- Division of Pulmonary Medicine, The First Affiliated Hospital of Wenzhou Medical University and Key Laboratory of Heart and Lung, Wenzhou, Zhejiang 325035, P.R. China
| | - Xiaomei Xu
- Division of Pulmonary Medicine, The First Affiliated Hospital of Wenzhou Medical University and Key Laboratory of Heart and Lung, Wenzhou, Zhejiang 325035, P.R. China
| | - Xueding Cai
- Division of Pulmonary Medicine, The First Affiliated Hospital of Wenzhou Medical University and Key Laboratory of Heart and Lung, Wenzhou, Zhejiang 325035, P.R. China
| | - Liangxing Wang
- Division of Pulmonary Medicine, The First Affiliated Hospital of Wenzhou Medical University and Key Laboratory of Heart and Lung, Wenzhou, Zhejiang 325035, P.R. China
| | - Xiaoying Huang
- Division of Pulmonary Medicine, The First Affiliated Hospital of Wenzhou Medical University and Key Laboratory of Heart and Lung, Wenzhou, Zhejiang 325035, P.R. China
| |
Collapse
|
58
|
Nakanishi A, Minami A, Kitagishi Y, Ogura Y, Matsuda S. BRCA1 and p53 tumor suppressor molecules in Alzheimer's disease. Int J Mol Sci 2015; 16:2879-92. [PMID: 25636033 PMCID: PMC4346871 DOI: 10.3390/ijms16022879] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2014] [Revised: 11/20/2014] [Accepted: 01/20/2015] [Indexed: 12/16/2022] Open
Abstract
Tumor suppressor molecules play a pivotal role in regulating DNA repair, cell proliferation, and cell death, which are also important processes in the pathogenesis of Alzheimer’s disease. Alzheimer’s disease is the most common neurodegenerative disorder, however, the precise molecular events that control the death of neuronal cells are unclear. Recently, a fundamental role for tumor suppressor molecules in regulating neurons in Alzheimer’s disease was highlighted. Generally, onset of neurodegenerative diseases including Alzheimer’s disease may be delayed with use of dietary neuro-protective agents against oxidative stresses. Studies suggest that dietary antioxidants are also beneficial for brain health in reducing disease-risk and in slowing down disease-progression. We summarize research advances in dietary regulation for the treatment of Alzheimer’s disease with a focus on its modulatory roles in BRCA1 and p53 tumor suppressor expression, in support of further therapeutic research in this field.
Collapse
Affiliation(s)
- Atsuko Nakanishi
- Department of Food Science and Nutrition, Nara Women's University, Kita-Uoya Nishimachi, Nara 630-8506, Japan.
| | - Akari Minami
- Department of Food Science and Nutrition, Nara Women's University, Kita-Uoya Nishimachi, Nara 630-8506, Japan.
| | - Yasuko Kitagishi
- Department of Food Science and Nutrition, Nara Women's University, Kita-Uoya Nishimachi, Nara 630-8506, Japan.
| | - Yasunori Ogura
- Department of Food Science and Nutrition, Nara Women's University, Kita-Uoya Nishimachi, Nara 630-8506, Japan.
| | - Satoru Matsuda
- Department of Food Science and Nutrition, Nara Women's University, Kita-Uoya Nishimachi, Nara 630-8506, Japan.
| |
Collapse
|
59
|
Zhang J, Liu S, Xu B, Li G, Li G, Huang A, Wu B, Peng L, Song M, Xie Q, Lin W, Xie W, Wen S, Zhang Z, Xu X, Liang S. Study of baicalin on sympathoexcitation induced by myocardial ischemia via P2X3 receptor in superior cervical ganglia. Auton Neurosci 2014; 189:8-15. [PMID: 25554221 DOI: 10.1016/j.autneu.2014.12.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2014] [Revised: 12/05/2014] [Accepted: 12/09/2014] [Indexed: 10/24/2022]
Abstract
After the myocardial ischemia, injured myocardial tissues released large quantity of ATP, which activated P2X3 receptor in superior cervical ganglia and made the SCG postganglionic neurons excited. Excitatory of sympathetic postganglionic efferent neurons increased the blood pressure and heart rates, which aggravated the myocardial ischemic injury. Baicalin has anti-inflammatory and anti-oxidant properties. Our study showed that baicalin reduced the incremental concentration of serum CK-MB, cTn-T, epinephrine and ATP, decreased the up-regulated expression levels of P2X3 mRNA and protein in SCG after MI, and then inhibited the sympathetic excitatory activity triggered by MI injury. These results indicated that baicalin acted on P2X3 receptor was involved in the transmission of sympathetic excitation after the myocardial ischemic injury. Baicalin might decrease sympathetic activity via inhibiting P2X3 receptor in rat SCG to protect the myocardium.
Collapse
Affiliation(s)
- Jun Zhang
- Department of Physiology, Medical School of Nanchang University, Nanchang 330006, PR China
| | - Shuangmei Liu
- Department of Physiology, Medical School of Nanchang University, Nanchang 330006, PR China
| | - Baohua Xu
- Department of Laboratory Animal, Medical School of Nanchang University, Nanchang 330006, PR China
| | - Guodong Li
- Department of Physiology, Medical School of Nanchang University, Nanchang 330006, PR China
| | - Guilin Li
- Department of Physiology, Medical School of Nanchang University, Nanchang 330006, PR China
| | - An Huang
- Jiangxi University of Finance and Economics, Nanchang, Jiangxi 330006, PR China
| | - Bing Wu
- Department of Physiology, Medical School of Nanchang University, Nanchang 330006, PR China
| | - Lichao Peng
- Department of Physiology, Medical School of Nanchang University, Nanchang 330006, PR China
| | - Miaomiao Song
- Department of Physiology, Medical School of Nanchang University, Nanchang 330006, PR China
| | - Qiuyu Xie
- 2012 Grade of Department of Clinical Medicine of Nanchang University, Nanchang 330006, PR China
| | - Weijian Lin
- 2012 Grade of Department of Clinical Medicine of Nanchang University, Nanchang 330006, PR China
| | - Wei Xie
- 2012 Grade of Department of Clinical Medicine of Nanchang University, Nanchang 330006, PR China
| | - Shiyao Wen
- 2012 Grade of Department of Clinical Medicine of Nanchang University, Nanchang 330006, PR China
| | - Zhedong Zhang
- 2012 Grade of Department of Clinical Medicine of Nanchang University, Nanchang 330006, PR China
| | - Xiaoling Xu
- Department of Biomedical Engineering, Information Engineering College of Nanchang University, Nanchang 330006, PR China
| | - Shangdong Liang
- Department of Physiology, Medical School of Nanchang University, Nanchang 330006, PR China; Institute of Life Science of Nanchang University, Nanchang 330006, PR China.
| |
Collapse
|
60
|
Tian J, Wang Y, Liu X, Zhou X, Li R. Rapamycin ameliorates IgA nephropathy via cell cycle-dependent mechanisms. Exp Biol Med (Maywood) 2014; 240:936-45. [PMID: 25349217 DOI: 10.1177/1535370214555666] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2014] [Accepted: 09/07/2014] [Indexed: 12/14/2022] Open
Abstract
IgA nephropathy is the most frequent type of glomerulonephritis worldwide. The role of cell cycle regulation in the pathogenesis of IgA nephropathy has been studied. The present study was designed to explore whether rapamycin ameliorates IgA nephropathy via cell cycle-dependent mechanisms. After establishing an IgA nephropathy model, rats were randomly divided into four groups. Coomassie Brilliant Blue was used to measure the 24-h urinary protein levels. Renal function was determined using an autoanalyzer. Proliferation was assayed via Proliferating Cell Nuclear Antigen (PCNA) immunohistochemistry. Rat mesangial cells were cultured and divided into the six groups. Methylthiazolyldiphenyl-tetrazolium bromide (MTT) and flow cytometry were used to detect cell proliferation and the cell cycle phase. Western blotting was performed to determine cyclin E, cyclin-dependent kinase 2, p27(Kip1), p70S6K/p-p70S6K, and extracellular signal-regulated kinase 1/2/p- extracellular signal-regulated kinase 1/2 protein expression. A low dose of the mammalian target of rapamycin (mTOR) inhibitor rapamycin prevented an additional increase in proteinuria, protected kidney function, and reduced IgA deposition in a model of IgA nephropathy. Rapamycin inhibited mesangial cell proliferation and arrested the cell cycle in the G1 phase. Rapamycin did not affect the expression of cyclin E and cyclin-dependent kinase 2. However, rapamycin upregulated p27(Kip1) at least in part via AKT (also known as protein kinase B)/mTOR. In conclusion, rapamycin can affect cell cycle regulation to inhibit mesangial cell proliferation, thereby reduce IgA deposition, and slow the progression of IgAN.
Collapse
Affiliation(s)
- Jihua Tian
- Department of Nephrology, The Affiliated People's Hospital of Shanxi Medical University, Shanxi Provincial People's Hospital, Shanxi Kidney Disease Institute, Taiyuan, Shanxi, 030012, China Department of Microbiology and Immunology, Shanxi Medical University, Taiyuan, Shanxi, 030001, China
| | - Yanhong Wang
- Department of Microbiology and Immunology, Shanxi Medical University, Taiyuan, Shanxi, 030001, China
| | - Xinyan Liu
- Department of Nephrology, The Affiliated People's Hospital of Shanxi Medical University, Shanxi Provincial People's Hospital, Shanxi Kidney Disease Institute, Taiyuan, Shanxi, 030012, China
| | - Xiaoshuang Zhou
- Department of Nephrology, The Affiliated People's Hospital of Shanxi Medical University, Shanxi Provincial People's Hospital, Shanxi Kidney Disease Institute, Taiyuan, Shanxi, 030012, China
| | - Rongshan Li
- Department of Nephrology, The Affiliated People's Hospital of Shanxi Medical University, Shanxi Provincial People's Hospital, Shanxi Kidney Disease Institute, Taiyuan, Shanxi, 030012, China
| |
Collapse
|
61
|
Luo Y, Zhang B, Dong HY, Liu Y, Li ZC, Dong MQ, Gao YQ. Prevention of hypoxic pulmonary hypertension by hypoxia-inducible expression of p27 in pulmonary artery smooth muscle cells. Gene Ther 2014; 21:751-758. [PMID: 24871579 DOI: 10.1038/gt.2014.49] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2013] [Revised: 03/25/2014] [Accepted: 04/11/2014] [Indexed: 11/08/2022]
Abstract
Hypoxia-induced proliferation of pulmonary artery smooth muscle cells (SMCs) is important in the development of hypoxic pulmonary hypertension (HPH). We constructed a lentivirial vector containing a smooth muscle-specific promoter and six copies of hypoxia response element to co-drive the expression of p27, the key cyclin-dependent kinase inhibitor that blocks the G1 to S phase transition in cell cycle progression, in pulmonary artery SMCs in hypoxia. Then in vivo we examined the prevention effects of the vector on HPH in mice and in vitro the specificity on the hypoxia-inducible expression of p27 in pulmonary artery SMCs. Hypobaric hypoxia for 4 weeks resulted in significant increases in the right ventricular systolic pressure, the ratio of right ventricle to left ventricle plus septal weight and the muscularization of pulmonary vessels in mice. Administration of the vector before hypoxia significantly prevented the effects of hypoxia. In vitro, the vector exhibited hypoxic inducibility and relatively specific expression in pulmonary artery SMCs, inhibited the hypoxia-induced proliferation of pulmonary artery SMCs and arrested more cells at G0/G1 phase. These results demonstrate that the hypoxia-inducible p27 expression prevents the development of HPH in mice.
Collapse
Affiliation(s)
- Y Luo
- 1] Department of Pathophysiology and High Altitude Physiology, Third Military Medical University, Chongqing, China [2] Key Laboratory of High Altitude Medicine, Ministry of Education, Third Military Medical University, Chongqing, China [3] Key Laboratory of High Altitude Medicine, PLA, Third Military Medical University, Chongqing, China [4] Department of Pathology and Pathophysiology, Fourth Military Medical University, Xi'an, China [5] Lung Injury and Repair Center, Fourth Military Medical University, Xi'an, China
| | - B Zhang
- 1] Department of Pathology and Pathophysiology, Fourth Military Medical University, Xi'an, China [2] Lung Injury and Repair Center, Fourth Military Medical University, Xi'an, China
| | - H-Y Dong
- 1] Department of Pathology and Pathophysiology, Fourth Military Medical University, Xi'an, China [2] Lung Injury and Repair Center, Fourth Military Medical University, Xi'an, China
| | - Y Liu
- 1] Department of Pathology and Pathophysiology, Fourth Military Medical University, Xi'an, China [2] Lung Injury and Repair Center, Fourth Military Medical University, Xi'an, China
| | - Z-C Li
- 1] Department of Pathology and Pathophysiology, Fourth Military Medical University, Xi'an, China [2] Lung Injury and Repair Center, Fourth Military Medical University, Xi'an, China
| | - M-Q Dong
- 1] Department of Pathology and Pathophysiology, Fourth Military Medical University, Xi'an, China [2] Lung Injury and Repair Center, Fourth Military Medical University, Xi'an, China
| | - Y-Q Gao
- 1] Department of Pathophysiology and High Altitude Physiology, Third Military Medical University, Chongqing, China [2] Key Laboratory of High Altitude Medicine, Ministry of Education, Third Military Medical University, Chongqing, China [3] Key Laboratory of High Altitude Medicine, PLA, Third Military Medical University, Chongqing, China
| |
Collapse
|
62
|
Suzuki M, Minami A, Nakanishi A, Kobayashi K, Matsuda S, Ogura Y, Kitagishi Y. Atherosclerosis and tumor suppressor molecules (review). Int J Mol Med 2014; 34:934-40. [PMID: 25069568 DOI: 10.3892/ijmm.2014.1866] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2014] [Accepted: 07/18/2014] [Indexed: 11/06/2022] Open
Abstract
Atherosclerosis, the major cause of heart attack and stroke, is a chronic inflammatory disease characterized by the formation of atherosclerotic plaque. Oxidized low-density lipoprotein through increased oxidative stress has been identified as one of the primary factors responsible for atherogenesis. Cell proliferation and death are key processes in the progression of atherosclerosis. The oxidative environment in areas of lipid accumulation is mainly created by the production of reactive oxygen species, which are assumed to mediate vascular tissue injury. Oxidative DNA damage and levels of DNA repair are reduced during dietary lipid lowering. The tumor suppressor molecules play a pivotal role in regulating cell proliferation, DNA repair and cell death, which are important processes in regulating the composition of atherosclerotic plaque. Accordingly, in this review, we discuss the fundamental role of tumor suppressor molecules in regulating atherogenesis. In particular, we discuss how tumor suppressor molecules are activated in the complex environment of atherosclerotic plaque, and regulate growth arrest, cell senescence and the apoptosis of vascular smooth muscle cells, which may protect against the progression of atherosclerosis. In addition, we discuss promising alternatives to the use of medications (such as statin) against atherosclerosis, namely diet, with the use of plant-derived supplements to modulate the expression and/or activity of tumor suppressor molecules. We also summarize the progress of research made on herbs with a focus on the modulatory roles of tumor suppressors, and on the molecular mechanisms underlying the prevention if atherosclerosis, supporting designs for further research in this field.
Collapse
Affiliation(s)
- Miho Suzuki
- Department of Food Science and Nutrition, Nara Women's University, Nishimachi, Nara 630-8506, Japan
| | - Akari Minami
- Department of Food Science and Nutrition, Nara Women's University, Nishimachi, Nara 630-8506, Japan
| | - Atsuko Nakanishi
- Department of Food Science and Nutrition, Nara Women's University, Nishimachi, Nara 630-8506, Japan
| | - Keiko Kobayashi
- Department of Food Science and Nutrition, Nara Women's University, Nishimachi, Nara 630-8506, Japan
| | - Satoru Matsuda
- Department of Food Science and Nutrition, Nara Women's University, Nishimachi, Nara 630-8506, Japan
| | - Yasunori Ogura
- Department of Food Science and Nutrition, Nara Women's University, Nishimachi, Nara 630-8506, Japan
| | - Yasuko Kitagishi
- Department of Food Science and Nutrition, Nara Women's University, Nishimachi, Nara 630-8506, Japan
| |
Collapse
|
63
|
Baicalin inhibits hypoxia-induced pulmonary artery smooth muscle cell proliferation via the AKT/HIF-1α/p27-associated pathway. Int J Mol Sci 2014; 15:8153-68. [PMID: 24821539 PMCID: PMC4057725 DOI: 10.3390/ijms15058153] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2014] [Revised: 04/27/2014] [Accepted: 04/30/2014] [Indexed: 12/30/2022] Open
Abstract
Baicalin, a flavonoid compound purified from the dry roots of Scutellaria baicalensis Georgi, has been shown to possess various pharmacological actions. Previous studies have revealed that baicalin inhibits the growth of cancer cells through the induction of apoptosis. Pulmonary arterial hypertension (PAH) is a devastating disease characterized by enhanced pulmonary artery smooth muscle cell (PASMCs) proliferation and suppressed apoptosis. However, the potential mechanism of baicalin in the regulation of PASMC proliferation and the prevention of cardiovascular diseases remains unexplored. To test the effects of baicalin on hypoxia, we used rats treated with or without baicalin (100 mg·kg−1 each rat) at the beginning of the third week after hypoxia. Hemodynamic and pulmonary pathomorphology data showed that right ventricular systolic pressures (RVSP), the weight of the right ventricle/left ventricle plus septum (RV/LV + S) ratio and the medial width of pulmonary arterioles were much higher in chronic hypoxia. However, baicalin treatment repressed the elevation of RVSP, RV/LV + S and attenuated the pulmonary vascular structure remodeling (PVSR) of pulmonary arterioles induced by chronic hypoxia. Additionally, baicalin (10 and 20 μmol·L−1) treatment suppressed the proliferation of PASMCs and attenuated the expression of hypoxia-inducible factor-α (HIF-α) under hypoxia exposure. Meanwhile, baicalin reversed the hypoxia-induced reduction of p27 and increased AKT/protein kinase B phosphorylation p-AKT both in vivo and in vitro. These results suggested that baicalin could effectively attenuate PVSR and hypoxic pulmonary hypertension.
Collapse
|
64
|
Nayak MK, Agrawal AS, Bose S, Naskar S, Bhowmick R, Chakrabarti S, Sarkar S, Chawla-Sarkar M. Antiviral activity of baicalin against influenza virus H1N1-pdm09 is due to modulation of NS1-mediated cellular innate immune responses. J Antimicrob Chemother 2014; 69:1298-310. [DOI: 10.1093/jac/dkt534] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
|
65
|
Guo J, Li L, Wu YJ, Yan Y, Xu XN, Wang SB, Yuan TY, Fang LH, Du GH. Inhibitory Effects of Brazilin on the Vascular Smooth Muscle Cell Proliferation and Migration Induced by PDGF-BB. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2013; 41:1283-96. [DOI: 10.1142/s0192415x13500869] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Abnormal vascular smooth muscle cell (VSMC) proliferation and migration contribute to the pathogenesis of vascular diseases including atherosclerosis and restenosis. Brazilin isolated from the heartwood of Caesalpinia sappan L. has been reported to exhibit various biological activities, such as anti-platelet aggregation, anti-inflammation, vasorelaxation and pro-apoptosis. However, the functional effects of Brazilin on VSMCs remain unexplored. The present study investigated the potential effects of Brazilin on platelet-derived growth factor (PDGF)-BB induced VSMC proliferation and migration as well as the underlying mechanism of action. VSMC proliferation and migration were measured by Crystal Violet Staining, wound-healing and Boyden chamber assays, respectively. Cell cycle was analyzed by flow cytometry. Enzymatic action of matrix metalloproteinase-9 (MMP-9) was carried out by gelatin zymography. Expression of adhesion molecules, cell cycle regulatory proteins, the phosphorylated levels of PDGF receptor β (PDGF-Rβ), Src, extracellular signal regulated kinase (ERK) and Akt were tested by immunoblotting. The present study demonstrated that pretreatment with Brazilin dose-dependently inhibited PDGF-BB stimulated VSMC proliferation and migration, which were associated with a cell-cycle arrest at G0/G1 phase, a reduction in the adhesion molecule expression and MMP-9 activation in VSMCs. Furthermore, the increase in PDGF-Rβ, Src, ERK1/2 and Akt phosphorylation induced by PDGF-BB were suppressed by Brazilin. These findings indicate that Brazilin inhibits PDGF-BB induced VSMC proliferation and migration, and the inhibitory effects of Brazilin may be associated with the blockade of PDGF-Rβ - ERK1/2 and Akt signaling pathways. In conclusion, the present study implicates that Brazilin may be useful as an anti-proliferative agent for the treatment of vascular diseases.
Collapse
Affiliation(s)
- Jing Guo
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Beijing 100050, China
| | - Li Li
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Beijing 100050, China
| | - Yu-Jie Wu
- Beijing Key Laboratory of Drug Targets Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Yu Yan
- Beijing Key Laboratory of Drug Targets Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Xiao-Na Xu
- Beijing Key Laboratory of Drug Targets Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Shou-Bao Wang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Beijing 100050, China
| | - Tian-Yi Yuan
- Beijing Key Laboratory of Drug Targets Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Lian-Hua Fang
- Beijing Key Laboratory of Drug Targets Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Guan-Hua Du
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Beijing 100050, China
| |
Collapse
|
66
|
Li L, Liu D, Bu D, Chen S, Wu J, Tang C, Du J, Jin H. Brg1-dependent epigenetic control of vascular smooth muscle cell proliferation by hydrogen sulfide. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2013; 1833:1347-55. [DOI: 10.1016/j.bbamcr.2013.03.002] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/06/2012] [Revised: 03/02/2013] [Accepted: 03/03/2013] [Indexed: 12/19/2022]
|
67
|
Inhibitory Effects of Hwangryunhaedok-Tang in 3T3-L1 Adipogenesis by Regulation of Raf/MEK1/ERK1/2 Pathway and PDK1/Akt Phosphorylation. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2013; 2013:413906. [PMID: 23762131 PMCID: PMC3676921 DOI: 10.1155/2013/413906] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/22/2013] [Accepted: 05/07/2013] [Indexed: 01/31/2023]
Abstract
Hwangryunhaedok-tang (HRT) has been long used as traditional medicine in Asia. However, inhibitory role of HRT is unclear in early stage of 3T3-L1 adipocyte differentiation related to signaling. In the present study, we investigated the inhibitory effects of HRT on upstream signaling of peroxisome proliferation-activity receptor-γ (PPAR-γ) and CCAAT/enhancer binding protein-β (C/EBP-β) expression in differentiation of 3T3-L1 preadipocytes. We found that HRT significantly inhibited the adipocyte differentiation by downregulating several adipocyte-specific transcription factors including PPAR-γ, C/EBP-α, and C/EBP-β in 3T3-L1 preadipocytes. Furthermore, we observed that HRT markedly inhibited the differentiation media-mediated phosphorylation of Raf/extracellular mitogen-activated protein kinase 1 (MEK1)/signal-regulated protein kinase 1/2 (ERK1/2) and phosphorylation of phosphoinositide-dependent kinase 1 (PDK1)/Akt. These results indicate that anti-adipogenesis mechanism involves the downregulation of the major transcription factors of adipogenesis including PPAR-γ and C/EBP-α through inhibition of Raf/MEK1/ERK1/2 phosphorylation and PDK1/Akt phosphorylation by HRT. Furthermore, high performance liquid chromatography (HPLC) analysis showed HRT contains active antiobesity constituents such as palmatine, berberine, geniposide, baicalin, baicalein, and wogonin. Taken together, this study suggested that anti-adipogenesis effects of HRT were accounted by downregulation of Raf/MEK1/ERK1/2 pathway and PDK1/Akt pathway during 3T3-L1 adipocyte differentiation.
Collapse
|
68
|
Choi BK, Cha BY, Yagyu T, Woo JT, Ojika M. Sponge-derived acetylenic alcohols, petrosiols, inhibit proliferation and migration of platelet-derived growth factor (PDGF)-induced vascular smooth muscle cells. Bioorg Med Chem 2013; 21:1804-10. [DOI: 10.1016/j.bmc.2013.01.039] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2012] [Revised: 01/16/2013] [Accepted: 01/18/2013] [Indexed: 10/27/2022]
|
69
|
Kitagishi Y, Kobayashi M, Matsuda S. Protection against Cancer with Medicinal Herbs via Activation of Tumor Suppressor. JOURNAL OF ONCOLOGY 2012; 2012:236530. [PMID: 23213333 PMCID: PMC3508586 DOI: 10.1155/2012/236530] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/13/2012] [Revised: 10/14/2012] [Accepted: 10/27/2012] [Indexed: 01/11/2023]
Abstract
Cancer remains a major cause of death, although research is ongoing for the development of more effective drugs. Some herbs have shown potential in preventing the occurrence and/or progression of cancer and other chronic diseases. They are being screened comprehensively to explore the possibility of development of feasible anticancer drugs. However, more information is required about the response to and the molecular target for specific herbs. It seems that there is a relationship between some medicinal herbs and tumor suppressor molecules which protect a cell from cancer. In this paper, we summarize the progress of recent research on herbs, with a particular focus on its anticancer role and molecular mechanisms underlying the cancer prevention property, supporting design for further research in this field.
Collapse
Affiliation(s)
- Yasuko Kitagishi
- Department of Environmental Health Science, Nara Women's University, Kita-Uoya Nishimachi, Nara 630-8506, Japan
| | | | | |
Collapse
|
70
|
Sahebkar A. Baicalin as a potentially promising drug for the management of sulfur mustard induced cutaneous complications: a review of molecular mechanisms. Cutan Ocul Toxicol 2012; 31:226-234. [PMID: 22107027 DOI: 10.3109/15569527.2011.633950] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Sulfur mustard (SM) is a bifunctional alkylating agent with strong blistering, irritant, mutagenic and cytotoxic properties. SM has been widely deployed as a chemical warfare agent for over a century, leading to extensive casualties. Skin is among the first and most heavily damaged organs upon SM exposure. Unfortunately, a considerable fraction of SM-intoxicated patients are still suffering from chronic cutaneous complications. While these complications adversely affect patients' quality of life, there is as yet no ideal treatment for them and therapeutic options are limited and mainly symptomatic. During recent decades, remarkable progress has been made in understanding molecular mechanisms underlying SM-induced dermatotoxicity and several intra- and extracellular targets have been identified. This review argues that baicalin, a bioactive flavonoid from the roots of Scutellaria spp., could counteract different molecular and biochemical abnormalities that mediate SM dermatotoxicity and could therefore be regarded as a promising therapeutic option for the management of SM-induced cutaneous lesions.
Collapse
Affiliation(s)
- Amirhossein Sahebkar
- Biotechnology Research Center and School of Pharmacy, Mashhad University of Medical Sciences (MUMS), Mashhad, Iran.
| |
Collapse
|
71
|
Joung H, Kwon JS, Kim JR, Shin S, Kang W, Ahn Y, Kook H, Kee HJ. Enhancer of polycomb1 lessens neointima formation by potentiation of myocardin-induced smooth muscle differentiation. Atherosclerosis 2012; 222:84-91. [PMID: 22398275 DOI: 10.1016/j.atherosclerosis.2012.02.015] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2011] [Revised: 02/02/2012] [Accepted: 02/09/2012] [Indexed: 11/19/2022]
Abstract
OBJECTIVE Previously, we reported that enhancer of polycomb1 (Epc1) induces skeletal muscle differentiation through the serum response factor (SRF). Considering that SRF plays a critical role in vascular smooth muscle cell (VSMC) differentiation, we expected that Epc1 also works in VSMCs. Here we examined the effect of Epc1 on neointima formation after arterial balloon injury and the underlying mechanism. METHODS Epc1 expression was examined in carotid artery injury or VSMC models. Interaction with myocardin (Myocd), a master regulator of smooth muscle differentiation, was examined by immunoprecipitation or promoter analysis with smooth muscle (SM) 22α promoter. Finally, we investigated whether local delivery of Epc1 regulated neointimal formation after injury. RESULTS Epc1 expression was down-regulated during proliferation induced by platelet-derived growth factor BB, whereas it was upregulated during differentiation in VSMCs. Forced expression of Epc1 induced VSMC differentiation. Epc1 physically interacted with Myocd to synergistically activate SM22α promoter activity. Transient transfection of Epc1 enhanced the physical interaction between Myocd and SRF, whereas that interaction was reduced when A10 cells were treated with siRNA for Epc1. Local delivery of Epc1 significantly reduced neointima formation induced by balloon injury. CONCLUSIONS Our results indicate that Epc1 induces VSMC differentiation by interacting with Myocd to induce SRF-dependent smooth muscle genes. We propose that Epc1 acts as a novel negative regulator of neointima formation after carotid injury.
Collapse
Affiliation(s)
- Hosouk Joung
- Department of Pharmacology and Medical Research Center for Gene Regulation, Chonnam National University Medical School, Republic of Korea
| | | | | | | | | | | | | | | |
Collapse
|
72
|
Li XW, Hu CP, Wu WH, Zhang WF, Zou XZ, Li YJ. Inhibitory effect of calcitonin gene-related peptide on hypoxia-induced rat pulmonary artery smooth muscle cells proliferation: role of ERK1/2 and p27. Eur J Pharmacol 2012; 679:117-26. [PMID: 22306243 DOI: 10.1016/j.ejphar.2012.01.015] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2011] [Revised: 01/05/2012] [Accepted: 01/13/2012] [Indexed: 11/29/2022]
Abstract
Calcitonin gene-related peptide (CGRP) inhibits angiotensin II-induced proliferation of aortic smooth muscle cells via inactivation of extracellular signal-regulated protein kinase 1/2 (ERK1/2). ERK1/2 is necessary for the degradation or down-regulation of the cell cycle inhibitor p27, and is also crucial in mediating proliferation of pulmonary artery smooth muscle cells (PASMCs). Whether ERK1/2/p27 signal pathway is involved in CGRP-mediated pathogenesis of pulmonary hypertension and vascular remodeling remains unknown. Pulmonary hypertension was induced by hypoxia in rats, and capsaicin (50 mg/kg, s.c.) was used to deplete endogenous CGRP. Proliferation of cultured PASMCs was determined by BrdU incorporation method and flow cytometry. The expression/level of CGRP, p27, ERK1/2, c-fos and c-myc was analyzed by radioimmunoassay, immunohistochemistry, real-time PCR or Western blot. Sensory CGRP depletion by capsaicin exacerbated hypoxia-induced pulmonary hypertension in rats, as shown by an increase in right ventricle systolic pressure, mean pulmonary artery pressure and vascular hypertrophy, accompanied with decreased p27 expression and increased expression of phosphorylated ERK1/2, c-fos and c-myc. Exogenous application of CGRP significantly inhibited hypoxia-induced proliferation of PASMCs concomitantly with increased p27 expression and decreased expression of phosphorylated ERK1/2, c-fos and c-myc. These effects of CGRP were abolished in the presence of CGRP(8-37). Knockdown of p27 also reversed the inhibitory effect of CGRP on proliferation of PASMCs and expression of c-fos and c-myc, but not on ERK1/2 phosphorylation. These results suggest that CGRP inhibits hypoxia-induced proliferation of PASMCs via ERK1/2/p27/c-fos/c-myc pathway. Down-regulation of CGRP may contribute to remodeling of pulmonary arteries in hypoxia-induced pulmonary hypertension.
Collapse
Affiliation(s)
- Xian-Wei Li
- Department of Pharmacology, School of Pharmaceutical Sciences, Central South University, Changsha 410078, China
| | | | | | | | | | | |
Collapse
|
73
|
Jiang C. Translational medicine in China I: perspectives from Chinese physicians and scientists. SCIENCE CHINA-LIFE SCIENCES 2012; 54:1071-3. [PMID: 22227895 PMCID: PMC7099172 DOI: 10.1007/s11427-011-4260-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 11/20/2011] [Indexed: 12/11/2022]
Affiliation(s)
- ChengYu Jiang
- Department of Biochemistry and Molecular Biology, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100005 China
| |
Collapse
|
74
|
Chanakira A, Dutta R, Charboneau R, Barke R, Santilli SM, Roy S. Hypoxia differentially regulates arterial and venous smooth muscle cell proliferation via PDGFR-β and VEGFR-2 expression. Am J Physiol Heart Circ Physiol 2011; 302:H1173-84. [PMID: 22159994 DOI: 10.1152/ajpheart.00411.2011] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Despite intensive research studies, theories have yet to focus on the contribution of hypoxia to patency differences observed clinically between arterial vs. venous grafts. This study investigates the differential hypoxic response of smooth muscle cells (SMC) to hypoxia-derived endothelial cell (EC) growth factors. Initiation of SMC proliferation under hypoxia (<5% O(2)) occurred only after incubation with hypoxic endothelial cell-conditioned media (H-ECM). After the investigation of several possible growth factors in the H-ECM that may be responsible for SMC proliferation, the greatest difference was observed in vascular endothelial growth factor (VEGF-A) and platelet-derived growth factor homodimer B (PDGF-BB) expression. VEGF-A increased (2-fold) significantly (P < 0.05) in arterial-derived smooth muscle cells (ASMC) under hypoxia compared with venous-derived smooth muscle cells (VSMC), which showed no significant change. VSMC showed significant (P < 0.05) increase in VEGFR-2 expression under hypoxia compared with ASMC. Incubation with VEGFR-2-neutralizing antibody/PDGFR antagonist in VSMC before addition of H-ECM resulted in decreased proliferation. ASMC proliferation under hypoxia did not decrease during incubation with VEGFR-2-neutralizing antibody but did decrease upon PDGFR antagonist incubation. Current therapies focusing on treating intimal hyperplasia have negated the fact that combinational therapy might be required to combat induction of SMC proliferation. Clinically, therapy with PDGFR antagonists plus anti-VEGFR-2 may prove to be efficacious in managing SMC proliferation in venous-derived grafts.
Collapse
Affiliation(s)
- Alice Chanakira
- Dept. of Pharmacology, University of Minnesota, 11 6-125 Jackson Hall, 312 Church Street South East, Minneapolis, MN 55455, USA
| | | | | | | | | | | |
Collapse
|
75
|
Seo J, Lee HS, Ryoo S, Seo JH, Min BS, Lee JH. Tangeretin, a citrus flavonoid, inhibits PGDF-BB-induced proliferation and migration of aortic smooth muscle cells by blocking AKT activation. Eur J Pharmacol 2011; 673:56-64. [PMID: 22040922 DOI: 10.1016/j.ejphar.2011.10.011] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2011] [Revised: 09/29/2011] [Accepted: 10/08/2011] [Indexed: 11/17/2022]
Abstract
Tangeretin, a natural polymethoxylated flavone concentrated in the peel of citrus fruits, is known to have antiproliferative, antiinvasive, antimetastatic and antioxidant activities. However, the effect of tangeretin on vascular smooth muscle cells (VSMCs) is unknown. This study examined the effect of tangeretin on platelet-derived growth factor (PDGF)-BB-induced proliferation and migration of rat aortic smooth muscle cells (RASMCs) as well as its underlying mechanisms. Tangeretin significantly inhibited proliferation, DNA synthesis and migration of PDGF-BB-stimulated RASMCs without inducing cell death. Treatment with tangeretin-induced cell-cycle arrest in the G₀/G₁ phase was associated with down-regulation of cyclin D1 and cyclin E in addition to up-regulation of p27(kip1). We also showed that tangeretin inhibited PDGF-BB-induced phosphorylation of AKT, while it had no effect on the phosphorylation of phospholipase Cγ (PLCγ), PDGF receptor β-chain (PDGF-Rβ) and extracellular signal-regulated kinase 1/2 (ERK1/2), c-Jun N-terminal kinase (JNK) and p38 mitogen-activated protein kinases (MAPKs). An in vitro kinase assay revealed that tangeretin inhibited AKT activity in a dose-dependent manner. Moreover, treatment of LY294002, a phosphoinositide 3-kinase (PI3K) inhibitor, had similar effects than that of tangeretin on the expression of p27(kip1) and cyclin D1, as well as cell migration in PDFG-BB-stimulated RASMCs. Taken together, these findings suggest that tangeretin could suppress PDGF-BB-induced proliferation and migration of RASMCs through the suppression of PI3K/AKT signaling pathway, and may be a potential candidate for preventing or treating vascular diseases, such as atherosclerosis and restenosis.
Collapse
MESH Headings
- Animals
- Aorta/cytology
- Aorta/drug effects
- Aorta/metabolism
- Becaplermin
- Cell Movement/drug effects
- Cell Proliferation/drug effects
- DNA/biosynthesis
- Dose-Response Relationship, Drug
- Flavones/administration & dosage
- Flavones/pharmacology
- Male
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Phosphatidylinositol 3-Kinases/metabolism
- Proto-Oncogene Proteins c-akt/metabolism
- Proto-Oncogene Proteins c-sis/pharmacology
- Rats
- Rats, Sprague-Dawley
- Signal Transduction/drug effects
Collapse
Affiliation(s)
- Juhee Seo
- Department of Biochemistry, College of Natural Sciences, Kangwon National University, Chuncheon 200-701, Republic of Korea
| | | | | | | | | | | |
Collapse
|
76
|
Guo Y, Huo RJ, Yao JF. Effect of baicalin on the JAK-STAT3 signaling pathway in human hepatocellular carcinoma cell line SMMC-7721. Shijie Huaren Xiaohua Zazhi 2011; 19:2363-2367. [DOI: 10.11569/wcjd.v19.i22.2363] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the effect of baicalin on the JAK-STAT3 signaling pathway in human hepatocellular carcinoma cell line SMMC-7721.
METHODS: SMMC-7721 cells were divided into four groups: blank control group, baicalin group, AG490 group, and baicalin plus AG490 group. The mRNA expressional level of STAT3 in SMMC-7721 cells was measured by RT-PCR. The protein expression levels of STAT3 and p-STAT3 in SMMC-7721 cells were measured by Western blot.
RESULTS: The expressional level of STAT3 mRNA in SMMC-7721 cells was significantly lower in the baicalin group than in the blank control group (0.505 ± 0.111 vs 0.697 ± 0.145, P < 0.05). The protein expressional levels of STAT3 and p-STAT3 in SMMC-7721 cells were also significantly lower in the baicalin group than in the blank control group (0.879 ± 0.012 vs 1.087 ± 0.015, 0.983 ± 0.085 vs 1.103 ± 0.074, both P < 0.05). Compared to the baicalin group, the protein expressional level of p-STAT3 in SMMC-7721 cells was significantly decreased in the baicalin plus AG490 (0.756 ± 0.103 vs 0.983 ± 0.085, P < 0.05).
CONCLUSION: Baicalin could significantly down-regulate the mRNA and protein expressional levels of STAT3 and p-STAT3 and inhibit the activation of STAT3 protein in SMMC-7721 cells.
Collapse
|
77
|
Farooqi AA, Waseem S, Riaz AM, Dilawar BA, Mukhtar S, Minhaj S, Waseem MS, Daniel S, Malik BA, Nawaz A, Bhatti S. PDGF: the nuts and bolts of signalling toolbox. Tumour Biol 2011; 32:1057-70. [PMID: 21769672 DOI: 10.1007/s13277-011-0212-3] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2011] [Accepted: 07/07/2011] [Indexed: 12/16/2022] Open
Abstract
PDGF is a growth factor and is extensively involved in multi-dimensional cellular dynamics. It switches on a plethora of molecules other than its classical pathway. It is engaged in various transitions of development; however, if the unleashed potentials lead astray, it brings forth tumourigenesis. Conventionally, it has been assumed that the components of this signalling pathway show fidelity and act with a high degree of autonomy. However, as illustrated by the PDGF signal transduction, reinterpretation of recent data suggests that machinery is often shared between multiple pathways, and other components crosstalk to each other through multiple mechanisms. It is important to note that metastatic cascade is an intricate process that we have only begun to understand in recent years. Many of the early steps of this PDGF cascade are not readily targetable in the clinic. In this review, we will unravel the paradoxes with reference to mitrons and cellular plasticity and discuss how disruption of signalling cascade triggers cellular proliferation phase transition and metastasis. We will also focus on the therapeutic interventions to counteract resultant molecular disorders.
Collapse
Affiliation(s)
- Ammad Ahmad Farooqi
- Institute of Molecular Biology and Biotechnology (IMBB), The University of Lahore, 1 km defence road, Lahore, Pakistan.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
78
|
Chan JY, Koon JC, Leung PC, Che CT, Fung KP. Suppression of low-density lipoprotein oxidation, vascular smooth muscle cell proliferation and migration by a herbal extract of Radix Astragali, Radix Codonopsis and Cortex Lycii. Altern Ther Health Med 2011; 11:32. [PMID: 21513503 PMCID: PMC3107819 DOI: 10.1186/1472-6882-11-32] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2010] [Accepted: 04/22/2011] [Indexed: 11/10/2022]
Abstract
Background Atherosclerosis is a major cause of death in developed world. Atherosclerosis is characterized by low-density lipoprotein deposition in the arterial wall which ultimately begets the formation of lesions. Rupture of lesions finally leads to clinical events such as heart attack and stroke. Atherosclerosis is a complication associated with diabetes. In patients with diabetes, the risk of atherosclerosis is three to five folds greater than in non-diabetics. Our previous study showed that a herbal extract of Radix Astragali, Radix Codonopsis and Cortex Lycii, namely SR10, could improve glucose homeostasis both in vitro and in vivo. In this study, we want to further investigate the efficacy of SR10 in treating atherosclerosis. Method The inhibitory effect of SR10 on low-density lipoprotein oxidation was investigated using free radical-induced erythrocyte hemolysis model and copper ion-induced low-density lipoprotein oxidation model. Since vascular smooth muscle cell proliferation and migration are important processes in atherogenesis, we also examined the effect of SR10 in inhibiting these events. Results Our results showed that SR10 inhibited erythrocyte hemolysis with IC50 value at 0.25 mg/ml and significantly prolonged low-density lipoprotein oxidation in vitro. SR10 attenuated platelet derived growth factor-BB-induced vascular smooth muscle cell proliferation by promoting cell cycle arrest at G0/G1 phase as well as inhibiting vascular smooth muscle cell migration. Conclusion The potential application of SR10 in treating atherosclerosis has been implied in this study. Animal model will be needed to further verify the efficacy of SR10 in future.
Collapse
|
79
|
Weakley SM, Wang X, Mu H, Lü J, Lin PH, Yao Q, Chen C. Ginkgolide A-gold nanoparticles inhibit vascular smooth muscle proliferation and migration in vitro and reduce neointimal hyperplasia in a mouse model. J Surg Res 2011; 171:31-9. [PMID: 21571322 DOI: 10.1016/j.jss.2011.03.018] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2011] [Revised: 02/17/2011] [Accepted: 03/04/2011] [Indexed: 11/29/2022]
Abstract
BACKGROUND Neointimal formation is mediated by phenotypic changes in vascular smooth muscle cells (SMC) and is an important mediator of restenosis following arterial reconstruction. We conjugated antioxidant ginkgolide A (GA) to gold nanoparticles (GNP) to determine the effect of GA delivery on neointimal formation. MATERIALS AND METHODS GA was conjugated to 80 nm GNP in an overnight incubation. Mouse P53LMAC01 vascular SMC were treated with various doses of GA-GNP, GA alone, GNP alone, and no treatment control. Cell proliferation and migration were analyzed, and superoxide anion levels and the phosphorylation status of ERK1/2 were determined. Mice underwent ligation of the common carotid artery along with local treatment with GNP (control) or GA-GNP. The carotid artery was harvested and subjected to immunohistochemical analysis. RESULTS GA-GNP treatment significantly inhibited SMC proliferation and migration in vitro in comparison to GNP treatment alone, and the effect persisted for up to 72 h after treatment. Treatment with GA-GNP also reduced superoxide anion levels in vitro. PDGF-BB substantially induced ERK1/2 phosphorylation in GNP control cells; this PDGE-BB induced ERK1/2 phosphorylation was significantly inhibited in GA-GNP-treated cells compared with GNP only. GA-GNP significantly reduced neointimal hyperplasia after injury in mice, and proliferating cell nuclear antigen (PCNA) staining was reduced substantially in the arteries of mice treated with GA-GNP. CONCLUSIONS GA-GNP reduce vascular SMC proliferation and migration in vitro through reduced activation of ERK1/2. Local treatment with GA-GNP in areas of arterial injury reduced neointimal hyperplasia and subsequent stenosis.
Collapse
Affiliation(s)
- Sarah M Weakley
- Molecular Surgeon Research Center, Division of Vascular Surgery and Endovascular Therapy, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, Texas 77030, USA
| | | | | | | | | | | | | |
Collapse
|