1
|
Iori S, D'Onofrio C, Laham-Karam N, Mushimiyimana I, Lucatello L, Montanucci L, Lopparelli RM, Bonsembiante F, Capolongo F, Pauletto M, Dacasto M, Giantin M. Generation and characterization of cytochrome P450 3A74 CRISPR/Cas9 knockout bovine foetal hepatocyte cell line (BFH12). Biochem Pharmacol 2024; 224:116231. [PMID: 38648904 DOI: 10.1016/j.bcp.2024.116231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 04/04/2024] [Accepted: 04/19/2024] [Indexed: 04/25/2024]
Abstract
In human, the cytochrome P450 3A (CYP3A) subfamily of drug-metabolizing enzymes (DMEs) is responsible for a significant number of phase I reactions, with the CYP3A4 isoform superintending the hepatic and intestinal metabolism of diverse endobiotic and xenobiotic compounds. The CYP3A4-dependent bioactivation of chemicals may result in hepatotoxicity and trigger carcinogenesis. In cattle, four CYP3A genes (CYP3A74, CYP3A76, CYP3A28 and CYP3A24) have been identified. Despite cattle being daily exposed to xenobiotics (e.g., mycotoxins, food additives, drugs and pesticides), the existing knowledge about the contribution of CYP3A in bovine hepatic metabolism is still incomplete. Nowadays, CRISPR/Cas9 mediated knockout (KO) is a valuable method to generate in vivo and in vitro models for studying the metabolism of xenobiotics. In the present study, we successfully performed CRISPR/Cas9-mediated KO of bovine CYP3A74, human CYP3A4-like, in a bovine foetal hepatocyte cell line (BFH12). After clonal expansion and selection, CYP3A74 ablation was confirmed at the DNA, mRNA, and protein level. The subsequent characterization of the CYP3A74 KO clone highlighted significant transcriptomic changes (RNA-sequencing) associated with the regulation of cell cycle and proliferation, immune and inflammatory response, as well as metabolic processes. Overall, this study successfully developed a new CYP3A74 KO in vitro model by using CRISPR/Cas9 technology, which represents a novel resource for xenobiotic metabolism studies in cattle. Furthermore, the transcriptomic analysis suggests a key role of CYP3A74 in bovine hepatocyte cell cycle regulation and metabolic homeostasis.
Collapse
Affiliation(s)
- Silvia Iori
- Department of Comparative Biomedicine and Food Science, University of Padua, Viale dell'Università 16, Legnaro, 35020 Padua, Italy
| | - Caterina D'Onofrio
- Department of Comparative Biomedicine and Food Science, University of Padua, Viale dell'Università 16, Legnaro, 35020 Padua, Italy
| | - Nihay Laham-Karam
- University of Eastern Finland, A.I. Virtanen Institute for Molecular Sciences, Neulaniementie 2, 70211 Kuopio, Finland
| | - Isidore Mushimiyimana
- University of Eastern Finland, A.I. Virtanen Institute for Molecular Sciences, Neulaniementie 2, 70211 Kuopio, Finland
| | - Lorena Lucatello
- Department of Comparative Biomedicine and Food Science, University of Padua, Viale dell'Università 16, Legnaro, 35020 Padua, Italy
| | - Ludovica Montanucci
- Department of Neurology, University of Texas Health Science Center, 6431 Fannin Street, Houston, TX, OH 44106, USA
| | - Rosa Maria Lopparelli
- Department of Comparative Biomedicine and Food Science, University of Padua, Viale dell'Università 16, Legnaro, 35020 Padua, Italy
| | - Federico Bonsembiante
- Department of Animal Medicine, Production and Health, University of Padua, Viale dell'Università 16, Legnaro, 35020 Padua, Italy
| | - Francesca Capolongo
- Department of Comparative Biomedicine and Food Science, University of Padua, Viale dell'Università 16, Legnaro, 35020 Padua, Italy
| | - Marianna Pauletto
- Department of Comparative Biomedicine and Food Science, University of Padua, Viale dell'Università 16, Legnaro, 35020 Padua, Italy
| | - Mauro Dacasto
- Department of Comparative Biomedicine and Food Science, University of Padua, Viale dell'Università 16, Legnaro, 35020 Padua, Italy
| | - Mery Giantin
- Department of Comparative Biomedicine and Food Science, University of Padua, Viale dell'Università 16, Legnaro, 35020 Padua, Italy.
| |
Collapse
|
2
|
Li X, Lin Z, Wang P, Zhou C, Xu J, Lin J, Lin D, Zhang D. Tetramethylpyrazine-Rhein Derivative inhibits the migration of canine inflammatory mammary carcinoma cells by mitochondrial damage-mediated apoptosis and cadherins downregulation. Biomed Pharmacother 2023; 162:114731. [PMID: 37086510 DOI: 10.1016/j.biopha.2023.114731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 04/10/2023] [Accepted: 04/17/2023] [Indexed: 04/24/2023] Open
Abstract
BACKGROUND Canine inflammatory mammary carcinoma (CIMC) has a high incidence of metastasis, high lethality, and poor prognosis, which needs novel adjuvant agents. Tetramethylpyrazine-Rhein Derivative (TRD) has been shown to have antitumor activity, which is a potential research direction for CIMC. PURPOSE This study evaluated the efficacy of TRD on CIMC in vitro and in vivo, and provided possibilities for the application of active compounds in traditional Chinese medicine. METHODS In vitro, TRD cytotoxicity was measured with CCK-8. Flow cytometry and transmission electron microscope were used to detect the cell cycle, cell death, and changes in mitochondria. Wound-healing assay, cell invasion assay, and scanning electron microscope were used to evaluate the suppression of cell migration and invasion. Expression changes were detected by RT-qPCR and western blot assay. In vivo, the lung metastasis models were randomly divided into control, low-dose TRD, high-dose TRD, and positive groups. Each group was administered orally once a day for 18 days and took in vivo imaging photos. RESULTS The IC50 of TRD in CHMp and MDCK were 42.59 and 79.37 μM, respectively. TRD mediated cell apoptosis by mitochondrial damage and caused S and G2/M phase arrest by downregulating cyclin B1. Moreover, TRD reduced filopodia and inhibited cell migration by downregulating cadherins. In CIMC lung metastasis models, TRD could effectively inhibit tumor growth (P < 0.001) in the lungs without significant toxicity. CONCLUSION TRD showed potential activity to inhibit CIMC lung metastasis with multi-target and low toxicity.
Collapse
Affiliation(s)
- Xinqiu Li
- College of Veterinary Medicine, China Agricultural University, Beijing 100193, PR China
| | - Zixiang Lin
- College of Veterinary Medicine, China Agricultural University, Beijing 100193, PR China
| | - Penglong Wang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, PR China
| | - Chaoyu Zhou
- College of Veterinary Medicine, China Agricultural University, Beijing 100193, PR China
| | - Jiawei Xu
- College of Veterinary Medicine, China Agricultural University, Beijing 100193, PR China
| | - Jiahao Lin
- College of Veterinary Medicine, China Agricultural University, Beijing 100193, PR China.
| | - Degui Lin
- College of Veterinary Medicine, China Agricultural University, Beijing 100193, PR China.
| | - Di Zhang
- College of Veterinary Medicine, China Agricultural University, Beijing 100193, PR China.
| |
Collapse
|
3
|
Liang R, Zhang G, Xu W, Liu W, Tang Y. Tetramethylpyrazine Inhibits the Proliferation and Invasion of Glioma Cells by Regulating the UBL7-AS1/miR-144-3p Pathway. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2022; 2022:5261285. [PMID: 36045665 PMCID: PMC9423964 DOI: 10.1155/2022/5261285] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 06/19/2022] [Accepted: 06/21/2022] [Indexed: 12/09/2022]
Abstract
This work aims to investigate the effects of tetramethylpyrazine (TMP) on the proliferation, migration, and invasion of glioma cells and to analyze the regulation mechanism of TMP on the long noncoding RNA UBL7-AS1/miR-144-3p pathway. Glioma cell line and normal astrocytes were collected. The expression of UBL7-AS1 was detected by real-time PCR. The glioma cells were overexpressed with UBL7-AS1. CCK-8 and Transwell assays were used to detect cell proliferation and cell invasion ability, respectively. Bioinformatics was adopted to predict the possible regulatory mechanisms of UBL7-AS1. The dual luciferase reporter gene was applied to verify the regulatory effect of RNA UBL7-AS1 with miR-144-3p. TMP inhibited the proliferation and invasion of glioma cells. UBL7-AS1 was highly expressed in glioma tissues and cells. The overexpression of UBL7-AS1 promotes the cell proliferation and invasion of glioma. UBL7-AS1 can act as a sponge for miR-144-3p in glioma cells. The overexpression of UBL7-AS1 can reverse the inhibition of TMP on proliferation, migration, and invasion of glioma cells. TMP inhibits the proliferation, migration, and invasion of glioma cells by regulating the UBL7-AS1/miR-144-3p pathway.
Collapse
Affiliation(s)
- Rui Liang
- Department of Neurosurgery, The First Hospital of Jiujiang City, Jiujiang, Jiangxi 332000, China
| | - Guofeng Zhang
- Department of Neurosurgery, The First Hospital of Jiujiang City, Jiujiang, Jiangxi 332000, China
| | - Wenhua Xu
- Department of Neurosurgery, The First Hospital of Jiujiang City, Jiujiang, Jiangxi 332000, China
| | - Weibing Liu
- Department of Neurosurgery, The First Hospital of Jiujiang City, Jiujiang, Jiangxi 332000, China
| | - Youjia Tang
- Department of Neurosurgery, The First Hospital of Jiujiang City, Jiujiang, Jiangxi 332000, China
| |
Collapse
|
4
|
Tetramethylpyrazine: A review on its mechanisms and functions. Biomed Pharmacother 2022; 150:113005. [PMID: 35483189 DOI: 10.1016/j.biopha.2022.113005] [Citation(s) in RCA: 67] [Impact Index Per Article: 33.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Revised: 04/15/2022] [Accepted: 04/19/2022] [Indexed: 11/21/2022] Open
Abstract
Ligusticum chuanxiong Hort (known as Chuanxiong in China, CX) is one of the most widely used and long-standing medicinal herbs in China. Tetramethylpyrazine (TMP) is an alkaloid and one of the active components of CX. Over the past few decades, TMP has been proven to possess several pharmacological properties. It has been used to treat a variety of diseases with excellent therapeutic effects. Here, the pharmacological characteristics and molecular mechanism of TMP in recent years are reviewed, with an emphasis on the signal-regulation mechanism of TMP. This review shows that TMP has many physiological functions, including anti-oxidant, anti-inflammatory, and anti-apoptosis properties; autophagy regulation; vasodilation; angiogenesis regulation; mitochondrial damage suppression; endothelial protection; reduction of proliferation and migration of vascular smooth muscle cells; and neuroprotection. At present, TMP is used in treating cardiovascular, nervous, and digestive system conditions, cancer, and other conditions and has achieved good curative effects. The therapeutic mechanism of TMP involves multiple targets, multiple pathways, and bidirectional regulation. TMP is, thus, a promising drug with great research potential.
Collapse
|
5
|
Ehata S, Miyazono K. Bone Morphogenetic Protein Signaling in Cancer; Some Topics in the Recent 10 Years. Front Cell Dev Biol 2022; 10:883523. [PMID: 35693928 PMCID: PMC9174896 DOI: 10.3389/fcell.2022.883523] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 05/09/2022] [Indexed: 12/19/2022] Open
Abstract
Bone morphogenetic proteins (BMPs), members of the transforming growth factor-β (TGF-β) family, are multifunctional cytokines. BMPs have a broad range of functions, and abnormalities in BMP signaling pathways are involved in cancer progression. BMPs activate the proliferation of certain cancer cells. Malignant phenotypes of cancer cells, such as increased motility, invasiveness, and stemness, are enhanced by BMPs. Simultaneously, BMPs act on various cellular components and regulate angiogenesis in the tumor microenvironment. Thus, BMPs function as pro-tumorigenic factors in various types of cancer. However, similar to TGF-β, which shows both positive and negative effects on tumorigenesis, BMPs also act as tumor suppressors in other types of cancers. In this article, we review important findings published in the recent decade and summarize the pro-oncogenic functions of BMPs and their underlying mechanisms. The current status of BMP-targeted therapies for cancers is also discussed.
Collapse
Affiliation(s)
- Shogo Ehata
- Department of Pathology, School of Medicine, Wakayama Medical University, Wakayama, Japan
- *Correspondence: Shogo Ehata,
| | - Kohei Miyazono
- Department of Applied Pathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
6
|
Yang S, Wu S, Dai W, Pang L, Xie Y, Ren T, Zhang X, Bi S, Zheng Y, Wang J, Sun Y, Zheng Z, Kong J. Tetramethylpyrazine: A Review of Its Antitumor Potential and Mechanisms. Front Pharmacol 2021; 12:764331. [PMID: 34975475 PMCID: PMC8716857 DOI: 10.3389/fphar.2021.764331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 11/18/2021] [Indexed: 11/13/2022] Open
Abstract
Cancer remains a major public health threat. The mitigation of the associated morbidity and mortality remains a major research focus. From a molecular biological perspective, cancer is defined as uncontrolled cell division and abnormal cell growth caused by various gene mutations. Therefore, there remains an urgent need to develop safe and effective antitumor drugs. The antitumor effect of plant extracts, which are characterized by relatively low toxicity and adverse effect, has attracted significant attention. For example, increasing attention has been paid to the antitumor effects of tetramethylpyrazine (TMP), the active component of the Chinese medicine Chuanqiong, which can affect tumor cell proliferation, apoptosis, invasion, metastasis, and angiogenesis, as well as reverse chemotherapeutic resistance in neoplasms, thereby triggering antitumor effects. Moreover, TMP can be used in combination with chemotherapeutic agents to enhance their effects and reduce the side effect associated with chemotherapy. Herein, we review the antitumor effects of TMP to provide a theoretical basis and foundation for the further exploration of its underlying antitumor mechanisms and promoting its clinical application.
Collapse
Affiliation(s)
- Shaojie Yang
- Biliary Surgery (2nd General) Unit, Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Shuodong Wu
- Biliary Surgery (2nd General) Unit, Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Wanlin Dai
- Innovation Institute of China Medical University, Shenyang, China
| | - Liwei Pang
- Biliary Surgery (2nd General) Unit, Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yaofeng Xie
- Department of Cardiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Tengqi Ren
- Biliary Surgery (2nd General) Unit, Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Xiaolin Zhang
- Biliary Surgery (2nd General) Unit, Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Shiyuan Bi
- Biliary Surgery (2nd General) Unit, Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yuting Zheng
- Biliary Surgery (2nd General) Unit, Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Jingnan Wang
- Biliary Surgery (2nd General) Unit, Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yang Sun
- Biliary Surgery (2nd General) Unit, Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Zhuyuan Zheng
- Biliary Surgery (2nd General) Unit, Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Jing Kong
- Biliary Surgery (2nd General) Unit, Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, China
- *Correspondence: Jing Kong,
| |
Collapse
|
7
|
Wen A, Zhu Y, Mazhar M, Qin L, Zeng H, Zhu Y. Enhancement of Anti-Proliferative Activity of the Extracts from Dehulled Adlay by Fermentation with Bacillus subtilis. Foods 2021; 10:foods10122959. [PMID: 34945511 PMCID: PMC8701002 DOI: 10.3390/foods10122959] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 11/25/2021] [Accepted: 11/29/2021] [Indexed: 01/18/2023] Open
Abstract
Dehulled adlay was fermented with Bacillus subtilis BJ3-2, the anti-proliferative activities of the extracts from fermented dehulled adlay were investigated with six types of tumor cells, and then the bioactive components and the anti-proliferative mechanism were primarily explored. Results showed that all the extracts of B.subtilis-fermented dehulled adlay (BDA) and dehulled adlay (DA) had no inhibition effect on human embryonic kidney 239T cells. The anti-proliferative activities of the extracts from BDA against six types of tumor cells were almost always significantly higher than DA. Compared with others, the n-butanol extract of BDA (BDA-Nb) exhibited stronger anti-proliferative activities against human leukemia K562 cells and human non-small cell lung cancer A549 cells. Importantly, the anti-proliferative activity of fermented dehulled adlay against K562 cells was firstly discovered. Meanwhile, BDA-Nb was rich in tetramethylpyrazine, γ-aminobutyric acid, protocatechuic, 2,3,4-trihydroxybenzoic, chlorogenic, p-hydroxybenzoic, caffeic, trans-cinnamic, ferulic acids, and rutin. BDA-Nb induced the proliferative inhibition of K562 and A549 cells due to abnormal cell morphology, the increased cell population in G1 phase and apoptosis rate, the downregulation of Bcl-2, and the upregulation of Bax and caspase-3/8/9. These results indicate that dehulled adlay fermented with B.subtilis could be a potential therapeutic agent for leukemia and lung cancer.
Collapse
Affiliation(s)
- Anyan Wen
- School of Liquor and Food Engineering, Guizhou University, Guiyang 550025, China; (A.W.); (Y.Z.); (H.Z.)
| | - Yong Zhu
- School of Liquor and Food Engineering, Guizhou University, Guiyang 550025, China; (A.W.); (Y.Z.); (H.Z.)
| | - Muhammad Mazhar
- College of Life Sciences, Guizhou University, Guiyang 550025, China;
| | - Likang Qin
- School of Liquor and Food Engineering, Guizhou University, Guiyang 550025, China; (A.W.); (Y.Z.); (H.Z.)
- Key Laboratory of Agricultural and Animal Products Storage and Processing of Guizhou Province, Guiyang 550025, China
- National & Local Joint Engineering Center for the Development and Utilization Technology of Drug and Food Resources in Southwest China, Guiyang 550025, China
- Correspondence:
| | - Haiying Zeng
- School of Liquor and Food Engineering, Guizhou University, Guiyang 550025, China; (A.W.); (Y.Z.); (H.Z.)
- Key Laboratory of Agricultural and Animal Products Storage and Processing of Guizhou Province, Guiyang 550025, China
| | - Yi Zhu
- Plant Protection and Plant Quarantine Station of Guizhou Province, Guiyang 550001, China;
| |
Collapse
|
8
|
Yang Q, Shen F, Zhang F, Bai X, Zhang Y, Zhang H. The combination of two natural medicines, Chuanxiong and Asarum: A review of the chemical constituents and pharmacological activities. JOURNAL OF CHEMICAL RESEARCH 2021. [DOI: 10.1177/17475198211039130] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Traditional Chinese medicine has been clinically used in China for many years, with experimental studies and clinical trials having demonstrated that it is safe and valid. Among many traditional natural medicines, Chuanxiong and Asarum have been proven to be effective in the treatment of relieving pain. Actually, as well as analgesic, they have common attributes, such as anti-inflammatory, cardiovascular benefits, and anticancer activities, with volatile oils being their major components. Furthermore, Chuanxiong and Asarum have been combined as drug pairs in the same prescription for thousands of years, with examples being Chuanxiong Chatiao San and Chuanxiongxixintang. More interestingly, their combination has better therapeutic effects on diseases than a single drug. After the combination of Chuanxiong and Asarum forms a blend, a series of changes take place in their chemical components, such as the contents of the main active ingredients, ferulic acid and ligustilide, increased significantly after this progress. At the same time, the pharmacological effects of the combination appearing to be more powerful, such as synergistic analgesic. This review focuses on the chemical constituents and pharmacological activities of Chuanxiong, Asarum, and Chuanxiong Asarum compositions.
Collapse
Affiliation(s)
- Qingcheng Yang
- College of Pharmacy, Dali University, Dali, P.R. China
- Department of Pharmacy, The First People’s Hospital of Kunming, Kunming, P.R. China
| | - Fangli Shen
- College of Pharmacy, Dali University, Dali, P.R. China
- Department of Pharmacy, The First People’s Hospital of Kunming, Kunming, P.R. China
| | - Fengqin Zhang
- College of Pharmacy, Dali University, Dali, P.R. China
| | - Xue Bai
- College of Pharmacy, Dali University, Dali, P.R. China
| | - Yanru Zhang
- College of Pharmacy, Dali University, Dali, P.R. China
| | - Haizhu Zhang
- College of Pharmacy, Dali University, Dali, P.R. China
- Western Yunnan Traditional Chinese Medicine and Ethnic Drug Engineering Center, Dali, P.R. China
| |
Collapse
|
9
|
Zhang N, Zhang S, Wu W, Lu W, Jiang M, Zheng N, Huang J, Wang L, Liu H, Zheng M, Wang J. Regorafenib inhibits migration, invasion, and vasculogenic mimicry of hepatocellular carcinoma via targeting ID1-mediated EMT. Mol Carcinog 2021; 60:151-163. [PMID: 33428809 DOI: 10.1002/mc.23279] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 12/22/2020] [Accepted: 12/28/2020] [Indexed: 12/20/2022]
Abstract
Regorafenib is approved for patients with unresectable hepatocellular carcinoma (HCC) following sorafenib. However, the effect of regorafenib on HCC metastasis and its mechanism are poorly understood. Here, our data showed that regorafenib significantly restrained the migration, invasion and vasculogenic mimicry (VM) of HCC cells, and downregulated the expression of epithelial-to-mesenchymal transition (EMT)/VM-related molecules. Using RNA-seq and cellular thermal shift assays, we found that inhibitor of differentiation 1 (ID1) was a key target of regorafenib. In HCC tissues, the protein expression of ID1 was positively correlated with EMT and VM formation (CD34- /PAS+ ). Functionally, ID1 knockdown inhibited HCC cell migration, invasion, metastasis, and VM formation in vitro and in vivo, with upregulation of E-cadherin and downregulation of Snail and VE-cadherin. Moreover, Snail overexpression promoted the migration, invasion, and VM formation of ID1 knockdown cells. Snail knockdown reduced the migration, invasion, and VM formation of ID1 overexpression cells. Finally, regorafenib suppressed VM formation and decreased the expression of ID1, VE-cadherin and Snail in HCC PDX model. In conclusion, we manifested that regorafenib distinctly inhibited EMT in HCC cells via targeting ID1, leading to the suppression of cell migration, invasion and VM formation. These findings suggest that regorafenib may be developed as a suitable therapeutic agent for HCC metastasis.
Collapse
Affiliation(s)
- Nan Zhang
- Fujian Key Laboratory for Translational Research in Cancer and Neurodegenerative Diseases, Institute for Translational Medicine, The School of Basic Medical Sciences, Fujian Medical University, Fujian, China
| | - Shaoqin Zhang
- Fujian Key Laboratory for Translational Research in Cancer and Neurodegenerative Diseases, Institute for Translational Medicine, The School of Basic Medical Sciences, Fujian Medical University, Fujian, China
| | - Wenda Wu
- Fujian Key Laboratory for Translational Research in Cancer and Neurodegenerative Diseases, Institute for Translational Medicine, The School of Basic Medical Sciences, Fujian Medical University, Fujian, China
| | - Wenxian Lu
- Fujian Key Laboratory for Translational Research in Cancer and Neurodegenerative Diseases, Institute for Translational Medicine, The School of Basic Medical Sciences, Fujian Medical University, Fujian, China
| | - Mingting Jiang
- Fujian Key Laboratory for Translational Research in Cancer and Neurodegenerative Diseases, Institute for Translational Medicine, The School of Basic Medical Sciences, Fujian Medical University, Fujian, China
| | - Ning Zheng
- Department of Pharmacology, Fujian Provincial Key Laboratory of Natural Medicine Pharmacology, The School of Pharmacy, Fujian Medical University, Fujian, China
| | - Jing Huang
- Fujian Key Laboratory for Translational Research in Cancer and Neurodegenerative Diseases, Institute for Translational Medicine, The School of Basic Medical Sciences, Fujian Medical University, Fujian, China
| | - Long Wang
- Fujian Key Laboratory for Translational Research in Cancer and Neurodegenerative Diseases, Institute for Translational Medicine, The School of Basic Medical Sciences, Fujian Medical University, Fujian, China
| | - Hekun Liu
- Fujian Key Laboratory for Translational Research in Cancer and Neurodegenerative Diseases, Institute for Translational Medicine, The School of Basic Medical Sciences, Fujian Medical University, Fujian, China
| | - Min Zheng
- Fujian Key Laboratory for Translational Research in Cancer and Neurodegenerative Diseases, Institute for Translational Medicine, The School of Basic Medical Sciences, Fujian Medical University, Fujian, China
| | - Jichuang Wang
- Fujian Key Laboratory for Translational Research in Cancer and Neurodegenerative Diseases, Institute for Translational Medicine, The School of Basic Medical Sciences, Fujian Medical University, Fujian, China
| |
Collapse
|
10
|
Chronic exposure to TNF reprograms cell signaling pathways in fibroblast-like synoviocytes by establishing long-term inflammatory memory. Sci Rep 2020; 10:20297. [PMID: 33219307 PMCID: PMC7679373 DOI: 10.1038/s41598-020-77380-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Accepted: 10/28/2020] [Indexed: 12/31/2022] Open
Abstract
Fibroblast-like synoviocytes (FLS) play a critical role in the pathogenesis of rheumatoid arthritis (RA). Chronic inflammation induces transcriptomic and epigenetic modifications that imparts a persistent catabolic phenotype to the FLS, despite their dissociation from the inflammatory environment. We analyzed high throughput gene expression and chromatin accessibility data from human and mouse FLS from our and other studies available on public repositories, with the goal of identifying the persistently reprogrammed signaling pathways driven by chronic inflammation. We found that the gene expression changes induced by short-term tumor necrosis factor-alpha (TNF) treatment were largely sustained in the FLS exposed to chronic inflammation. These changes that included both activation and repression of gene expression, were accompanied by the remodeling of chromatin accessibility. The sustained activated genes (SAGs) included established pro-inflammatory signaling components known to act at multiple levels of NF-kappaB, STAT and AP-1 signaling cascades. Interestingly, the sustained repressed genes (SRGs) included critical mediators and targets of the BMP signaling pathway. We thus identified sustained repression of BMP signaling as a unique constituent of the long-term inflammatory memory induced by chronic inflammation. We postulate that simultaneous targeting of these activated and repressed signaling pathways may be necessary to combat RA persistence.
Collapse
|
11
|
Pu ZH, Dai M, Xiong L, Peng C. Total alkaloids from the rhizomes of Ligusticum striatum: a review of chemical analysis and pharmacological activities. Nat Prod Res 2020; 36:3489-3506. [PMID: 33034219 DOI: 10.1080/14786419.2020.1830398] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Rhizome Chuanxiong (RCX), the dried rhizomes of Ligusticum striatum DC., is a geoauthentic TCM herb distributed in Sichuan province of China that possesses efficacy in promoting blood circulation, removing blood stasis and alleviating pain. Rhizome Chuanxiong total alkaloids (RCXTAs) are one of the major characteristic constituents of RCX with the effects of antimigraine, neuroprotective, cardioprotective and other cardiovascular and cerebrovascular diseases. Over the past years, rapid development of technology has advanced some aspects of RCXTAs. The aim of this review is to illustrate the recent advances in the chemical analysis and biological activities of RCXTAs, and to highlight new challenges.
Collapse
Affiliation(s)
- Zhong-Hui Pu
- Department of Laboratory Medicine, Chengdu Medical College, Chengdu, China
| | - Min Dai
- Department of Laboratory Medicine, Chengdu Medical College, Chengdu, China
| | - Liang Xiong
- Pharmacy College, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Cheng Peng
- Pharmacy College, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
12
|
Hu Z, Su H, Zeng Y, Lin C, Guo Z, Zhong F, Jiang K, Yuan G, He S. Tetramethylpyrazine ameliorates hepatic fibrosis through autophagy-mediated inflammation. Biochem Cell Biol 2020; 98:327-337. [PMID: 32383631 DOI: 10.1139/bcb-2019-0059] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Imbalanced immune response and hepatic fibrosis are key factors related to the progression of chronic liver diseases. Tetramethylpyrazine (TMP), a natural alkaloid, has been widely used for treating liver injury. In this study, we explored the effect of TMP on hepatic fibrosis and the related mechanisms regulating autophagy. METHODS A rat model of hepatic fibrosis and a model using an hepatic stellate cell line (HSC-T6) were created using CCl4 and platelet-derived growth factor (PDGF). Staining with haematoxylin and eosin (HE), Masson's stain, and TUNEL were performed for pathological diagnosis. ELISA, Western blotting, and immunofluorescence analyses were conducted to determine the expression levels of the specific markers for fibrosis, autophagy, inflammation, and signalling pathways. RESULTS TMP treatment significantly rescued pathological injury and hepatic fibrosis. It also alleviated imbalances in the immune system, accumulation of extracellular matrix, and autophagy signals in hepatic fibrosis. At the same time, we found that application of the autophagy inducer rapamycin enhanced the therapeutic effect of TMP, whereas the autophagy inhibitor 3-methyladenine, PI3K pathway inhibitor LY294002, and AKT pathway agonist SC79 did the opposite. CONCLUSIONS TMP exerts therapeutic effects in hepatic fibrosis mainly through promoting autophagy to ameliorate inflammation by inhibiting the AKT-mTOR signalling pathway, providing a new perspective for the treatment of chronic liver diseases.
Collapse
Affiliation(s)
- Zhigao Hu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, No. 6, Shuangyong Road, Nanning 530021, P.R. China.,Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, No. 6, Shuangyong Road, Nanning 530021, P.R. China
| | - Huizhao Su
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, No. 6, Shuangyong Road, Nanning 530021, P.R. China.,Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, No. 6, Shuangyong Road, Nanning 530021, P.R. China
| | - Yonglian Zeng
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, No. 6, Shuangyong Road, Nanning 530021, P.R. China.,Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, No. 6, Shuangyong Road, Nanning 530021, P.R. China
| | - Chengjie Lin
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, No. 6, Shuangyong Road, Nanning 530021, P.R. China.,Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, No. 6, Shuangyong Road, Nanning 530021, P.R. China
| | - Zhenya Guo
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, No. 6, Shuangyong Road, Nanning 530021, P.R. China.,Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, No. 6, Shuangyong Road, Nanning 530021, P.R. China
| | - Fudi Zhong
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, No. 6, Shuangyong Road, Nanning 530021, P.R. China.,Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, No. 6, Shuangyong Road, Nanning 530021, P.R. China
| | - Keqing Jiang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, No. 6, Shuangyong Road, Nanning 530021, P.R. China.,Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, No. 6, Shuangyong Road, Nanning 530021, P.R. China
| | - Guandou Yuan
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, No. 6, Shuangyong Road, Nanning 530021, P.R. China.,Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, No. 6, Shuangyong Road, Nanning 530021, P.R. China
| | - Songqing He
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, No. 6, Shuangyong Road, Nanning 530021, P.R. China.,Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, No. 6, Shuangyong Road, Nanning 530021, P.R. China
| |
Collapse
|
13
|
Zhou Y, Zhou Z, Ji Z, Yan W, Li H, Yu X. Tetramethylpyrazine reduces prostate cancer malignancy through inactivation of the DPP10‑AS1/CBP/FOXM1 signaling pathway. Int J Oncol 2020; 57:314-324. [PMID: 32319592 DOI: 10.3892/ijo.2020.5036] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2019] [Accepted: 02/06/2020] [Indexed: 11/05/2022] Open
Abstract
Tetramethylpyrazine (TMP), a Chinese herbal medicine, has been reported to possess anticancer effects. Emerging evidence suggests that various long noncoding RNAs (lncRNAs) serve important roles in cancer initiation and progression. In the present study, the tumor‑suppressive effects of TMP in human PCa cells was examined and the underlying mechanisms of its actions were determined. The data showed that TMP treatment reduced cell viability and increased apoptosis in a dose‑dependent manner. Reverse transcription‑quantitative PCR showed TMP treatment increased the expression of lncRNA DPP10‑AS1 in PCa cells. Furthermore, DPP10‑AS1 was also upregulated in TMP‑resistant PCa cells. Knockdown of DPP10‑AS1 reversed TMP resistance, whereas increased expression of DPP10‑AS1 abrogated the TMP‑mediated cytotoxicity in PCa cells. In addition, forkhead box M1 (FOXM1) was verified as the functional target of DPP10‑AS1, and knockdown of FOXM1 reversed the TMP/DPP10‑AS1‑induced cell cytotoxicity. Mechanistically, DPP10‑AS1 was associated with CREB binding protein, thereby induced H3K27ac enrichment at the promoter region of the FOXM1 gene. In conclusion, the present study showed that TMP may be a promising treatment agent for PCa and lncRNA DPP10‑AS1 may be a promising therapeutic target for TMP treatment.
Collapse
Affiliation(s)
- Yi Zhou
- Department of Urology, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100000, P.R. China
| | - Zhien Zhou
- Department of Urology, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100000, P.R. China
| | - Zhigang Ji
- Department of Urology, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100000, P.R. China
| | - Weigang Yan
- Department of Urology, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100000, P.R. China
| | - Hanzhong Li
- Department of Urology, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100000, P.R. China
| | - Xiao Yu
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100000, P.R. China
| |
Collapse
|
14
|
Lu L, Wang P, Zou Y, Zha Z, Huang H, Guan M, Wu Y, Liu G. IL-1β Promotes Stemness of Tumor Cells by Activating Smad/ID1 Signaling Pathway. Int J Med Sci 2020; 17:1257-1268. [PMID: 32547321 PMCID: PMC7294920 DOI: 10.7150/ijms.44285] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2020] [Accepted: 04/28/2020] [Indexed: 12/23/2022] Open
Abstract
Background: IL-1β is reported to be involved in cancer development and distant metastasis. However, the underlying mechanism of IL-1β upon malignant behaviors remains largely unknown. In this study, we aimed to study whether IL-1β could enhance the stemness traits of tumor cells. Methods: The concentrations of serum IL-1β in head and neck squamous cell carcinoma (HNSCC) and melanoma patients were detected using ELISA assay. The effect and mechanisms of IL-1β on tumor cell growth, migration, invasion and stemness characters were studied using HNSCC cell SCC7 and melanoma cell B16-F10. The underlying mechanisms were further explored. Results: Enhanced concentrations of IL-1β were positively correlated with advanced tumor stage in both HNSCC and melanoma patients. IL-1β treatment led to a significant increase in tumor growth both in vitro and in vivo. IL-1β stimulation promoted cell proliferation, colony formation and tumorigenicity. In addition, IL-1β-stimulated tumor cells gained enhanced capabilities on wounding healing and invasion capabilities. Moreover, IL-1β stimulation promoted the stem-like capabilities of both HNSCC cells and melanoma cells, including the enrichment of aldehyde dehydrogenase+ (ALDH+) cells, up-regulation of stem cell related markers Nanog, OCT4, and SOX2, sphere formation and chemoresistance. Mechanistically, IL-1β treatment promoted the phosphorylation of Smad1/5/8 and activated its downstream target inhibitor of differentiation 1 (ID1). Silencing ID1 abrogated sphere formation and upregulated expression of stemness genes which were induced by IL-1β stimulation. Conclusion: Our data demonstrates that IL-1β promotes the stemness of HNSCC and melanoma cells through activating Smad/ID1 signal pathway.
Collapse
Affiliation(s)
- Lin Lu
- Department of Medical Oncology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China, 510180.,Department of Medical Oncology, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China, 510180
| | - Peipei Wang
- Department of Medical Oncology, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China, 510180
| | - Yonghong Zou
- Department of Gynecology and Obstetrics, Ji'an City Center People's Hospital, Jiangxi, China, 343000
| | - Zhiqiang Zha
- Department of Medical Oncology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China, 510180
| | - Haowei Huang
- Department of Medical Oncology, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China, 510180
| | - Mingmei Guan
- Department of Medical Oncology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China, 510180.,Department of Medical Oncology, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China, 510180
| | - Yong Wu
- Department of Medical Oncology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China, 510180.,Department of Medical Oncology, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China, 510180
| | - Guolong Liu
- Department of Medical Oncology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China, 510180.,Department of Medical Oncology, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China, 510180
| |
Collapse
|
15
|
Zhao Z, Bo Z, Gong W, Guo Y. Inhibitor of Differentiation 1 (Id1) in Cancer and Cancer Therapy. Int J Med Sci 2020; 17:995-1005. [PMID: 32410828 PMCID: PMC7211148 DOI: 10.7150/ijms.42805] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Accepted: 03/20/2020] [Indexed: 02/07/2023] Open
Abstract
The inhibitor of DNA binding (Id) proteins are regulators of cell cycle and cell differentiation. Of all Id family proteins, Id1 is mostly linked to tumorigenesis, cellular senescence as well as cell proliferation and survival. Id1 is a stem cell-like gene more than a classical oncogene. Id1 is overexpressed in numerous types of cancers and exerts its promotion effect to these tumors through different pathways. Briefly, Id1 was found significantly correlated with EMT-related proteins, K-Ras signaling, EGFR signaling, BMP signaling, PI3K/Akt signaling, WNT and SHH signaling, c-Myc signaling, STAT3 signaling, RK1/2 MAPK/Egr1 pathway and TGF-β pathway, etc. Id1 has potent effect on facilitating tumorous angiogenesis and metastasis. Moreover, high expression of Id1 plays a facilitating role in the development of drug resistance, including chemoresistance, radiation resistance and resistance to drugs targeting angiogenesis. However, controversial results were also obtained. Overall, Id1 represent a promising target of anti-tumor therapeutics based on its potent promotion effect to cancer. Numerous drugs were found exerting their anti-tumor function through Id1-related signaling pathways, such as fucoidan, berberine, tetramethylpyrazine, crizotinib, cannabidiol and vinblastine.
Collapse
Affiliation(s)
- Zhengxiao Zhao
- Department of Oncology, the First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310006, China
| | - Zhiyuan Bo
- The Second Department of Biliary Tract Surgery, Shanghai Eastern Hepatobiliary Surgery Hospital, Shanghai 200438, China
| | - Weiyi Gong
- The Department of Integrative Medicine, Huashan Hospital, Fudan University, 12 Middle Urumqi Road, Shanghai 200040, PR China
| | - Yong Guo
- Department of Oncology, the First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310006, China
| |
Collapse
|
16
|
Qing L, Wu P, Zhou Z, Yu F, Tang J. Tetramethylpyrazine improved the survival of multiterritory perforator flaps by inducing angiogenesis and suppressing apoptosis via the Akt/Nrf2 pathway. DRUG DESIGN DEVELOPMENT AND THERAPY 2019; 13:1437-1447. [PMID: 31118578 PMCID: PMC6504674 DOI: 10.2147/dddt.s195090] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/18/2018] [Accepted: 03/06/2019] [Indexed: 12/15/2022]
Abstract
Background: Multiterritory perforator flaps were commonly designed to cover the large soft-tissue defects in reconstructive surgery. But the high risk of partial necrosis in the distal portion of the flaps hindered their clinical application. The purpose of this study was to evaluate the effects of tetramethylpyrazine (TMP) on the survival of the multiterritory perforator flaps and to explore the underlying mechanism. Materials and methods: Seventy-two Sprague–Dawley rats underwent multiterritory perforator flap procedure and were divided into three groups with 24 each. Flap survival and water content were measured, and the area of angiogenesis and apoptosis in the ischemia skin flaps were assessed on the postoperative day 7. The expressions of angiogenesis-related protein VEGF and apoptosis-related protein Bax, Bcl-2 in each group were detected by Western blotting, which also had been used to assess the expressions levels of Akt, p-Akt, and Nrf2. Results: Following TMP treatment, the survival area and number of microvessels presented in the skin flaps increased and tissue edema reduced on postoperative day 7. The expressions of angiogenesis-related protein VEGF increased in the TMP treatment group than in the control group. In addition, compared with the control group, TMP inhibited apoptosis, and increased the expression levels of p-Akt, Nrf2 in the areas of ischemia. These effects were reversed by an Akt protein inhibitor LY294002. Similarly, treatment with LY294002 inhibited TMP induced by interfering the Akt/Nrf2 signaling pathway. Conclusion: These results illustrated that TMP could promote the survival of multiterritory perforator flaps by enhancing angiogenesis and attenuating apoptosis. These were involved in Akt/Nrf2 signaling pathway.
Collapse
Affiliation(s)
- LiMing Qing
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China.,Department of Neurosurgery, Maryland University of Medicine School- Baltimore, Baltimore, MD, USA
| | - PanFeng Wu
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
| | - ZhengBing Zhou
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
| | - Fang Yu
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
| | - JuYu Tang
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
| |
Collapse
|
17
|
Gu S, Shen H, Zhou Y, Ni J, Zheng T, Mou Z, Hua X. Tetramethylpyrazine reduces the consequences of nitric oxide inhibition in pregnant rats. J Cell Physiol 2019; 234:19799-19806. [PMID: 30937928 DOI: 10.1002/jcp.28579] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Revised: 03/04/2019] [Accepted: 03/06/2019] [Indexed: 02/04/2023]
Abstract
Pre-eclampsia (PE) is closely associated with perinatal morbidity and mortality and we want to investigate tetramethylpyrazine (TMP)'s effects on PE. Pregnant Sprague-Dawley rats were randomly divided into five groups: normal pregnant (PC), PE, PE+TMP 20 mg/kg, PE+TMP 40 mg/kg, and PE+TMP 60 mg/kg group. The PE rat model was established via L-NAME treatment. Systolic blood pressures (SBP) and urinary protein concentration were detected via the tail-cuff method and CBB kit, respectively. mRNA levels of key genes were analyzed via quantitative PCR and protein levels of key genes were measured by ELISA or western blot. TMP decreased SBP and urinary protein concentration of PE rats. TMP inhibited L-NAME-induced decrease in pups alive ratio, pups weight, and the ratio of pups/placenta weight and reversed L-NAME induced changes in placental histology, whereas it had little effect on placental weight. Urinary nephrin and podocin expressions were enhanced and serum placental growth factor level was decreased in PE rats, whereas TMP inhibited the above phenomena. TMP suppressed L-NAME-induced sFlt-1 upregulation in serums and kidneys of PE rats, whereas it downregulated IL-6 and MCP-1 expression in PE rats' serums, placentas and kidneys. TMP also suppressed the increase in placental sFlt-1 and vascular endothelial growth factor level caused by L-NAME. In addition, TMP inhibited CHOP and GRP78 expressions and decreased the ratio of p-elF2α/elF2α in PE rats. TMP attenuated the consequences of NO inhibition in pregnant rats.
Collapse
Affiliation(s)
- Shengyi Gu
- Department of Obstetrics and Gynecology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Department of Obstetrics, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, China
| | - Huaxiang Shen
- Department of Obstetrics, Jiaxing Maternity and Child Health Hospital, Jiaxing, Zhejiang, China
| | - Yun Zhou
- Department of Obstetrics and Gynecology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Jiaying Ni
- Department of Obstetrics and Gynecology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Tao Zheng
- Department of Obstetrics and Gynecology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Zhengqian Mou
- Department of Obstetrics and Gynecology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Department of Obstetrics, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xiaolin Hua
- Department of Obstetrics, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
18
|
Zhang L, Ye Y, Long X, Xiao P, Ren X, Yu J. BMP signaling and its paradoxical effects in tumorigenesis and dissemination. Oncotarget 2018; 7:78206-78218. [PMID: 27661009 PMCID: PMC5363655 DOI: 10.18632/oncotarget.12151] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Accepted: 09/14/2016] [Indexed: 01/04/2023] Open
Abstract
Bone morphogenetic proteins (BMPs) play important roles in embryonic and postnatal development by regulating cell differentiation, proliferation, motility, and survival, thus maintaining homeostasis during organ and tissue development. BMPs can lead to tumorigenesis and regulate cancer progression in different stages. Therefore, we summarized studies on BMP expression, the clinical significance of BMP dysfunction in various cancer types, and the molecular regulation of various BMP-related signaling pathways. We emphasized on the paradoxical effects of BMPs on various aspects of carcinogenesis, including epithelial–mesenchymal transition (EMT), cancer stem cells (CSCs), and angiogenesis. We also reviewed the molecular mechanisms by which BMPs regulate tumor generation and progression as well as potential therapeutic targets against BMPs that might be valuable in preventing tumor growth and invasion.
Collapse
Affiliation(s)
- Lijie Zhang
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Caner, Key Laboratory of Cancer Prevention and Therapy, Clinical Research Center for Cancer, Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, P. R. China
| | - Yingnan Ye
- Cancer Molecular Diagnostic Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Caner, Key Laboratory of Cancer Prevention and Therapy, Clinical Research Center for Cancer, Tianjin, P. R. China
| | - Xinxin Long
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Caner, Key Laboratory of Cancer Prevention and Therapy, Clinical Research Center for Cancer, Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, P. R. China
| | - Pei Xiao
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Caner, Key Laboratory of Cancer Prevention and Therapy, Clinical Research Center for Cancer, Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, P. R. China
| | - Xiubao Ren
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Caner, Key Laboratory of Cancer Prevention and Therapy, Clinical Research Center for Cancer, Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, P. R. China
| | - Jinpu Yu
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Caner, Key Laboratory of Cancer Prevention and Therapy, Clinical Research Center for Cancer, Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, P. R. China.,Cancer Molecular Diagnostic Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Caner, Key Laboratory of Cancer Prevention and Therapy, Clinical Research Center for Cancer, Tianjin, P. R. China
| |
Collapse
|
19
|
Zhang F, Lu S, He J, Jin H, Wang F, Wu L, Shao J, Chen A, Zheng S. Ligand Activation of PPARγ by Ligustrazine Suppresses Pericyte Functions of Hepatic Stellate Cells via SMRT-Mediated Transrepression of HIF-1α. Am J Cancer Res 2018; 8:610-626. [PMID: 29344293 PMCID: PMC5771080 DOI: 10.7150/thno.22237] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2017] [Accepted: 10/22/2017] [Indexed: 12/31/2022] Open
Abstract
Rationale: Hepatic stellate cells (HSCs) are liver-specific pericytes regulating vascular remodeling during hepatic fibrosis. Here, we investigated how ligustrazine affects HSC pericyte functions. Methods: Rat HSC-T6 and human HSC-LX2 cells were cultured, and multiple molecular experiments including real-time PCR, Western blot, flow cytometry, immunofluorescence, electrophoretic mobility shift assay and co-immunoprecipitation were used to elucidate the underlying mechanisms. Molecular simulation and site-directed mutagenesis were performed to uncover the target molecule of ligustrazine. Rats were intoxicated with CCl4 for evaluating ligustrazine's effects in vivo. Results: Ligustrazine inhibited angiogenic cytokine production, migration, adhesion and contraction in HSCs, and activated PPARγ. Selective PPARγ inhibitor GW9662 potently abrogated ligustrazine suppression of HSC pericyte functions. Additionally, HIF-1α inhibitor PX-478 repressed HSC pericyte functions, and ligustrazine inhibited the transcription of HIF-1α, which was diminished by GW9662. Moreover, ligustrazine downregulation of HIF-1α was rescued by knockdown of SMRT, and ligustrazine increased PPARγ physical interaction with SMRT, which was abolished by GW9662. These findings collectively indicated that activation of PPARγ by ligustrazine led to transrepression of HIF-1α via a SMRT-dependent mechanism. Furthermore, molecular docking evidence revealed that ligustrazine bound to PPARγ in a unique double-molecule manner via hydrogen bonding with the residues Ser289 and Ser342. Site-directed mutation of Ser289 and/or Ser342 resulted in the loss of ligustrazine transrepression of HIF-1α in HSCs, indicating that interactions with both the residues were indispensable for ligustrazine effects. Finally, ligustrazine improved hepatic injury, angiogenesis and vascular remodeling in CCl4-induced liver fibrosis in rats. Conclusions: We discovered a novel ligand activation pattern for PPARγ transrepression of the target gene with therapeutic implications in HSC pericyte biology and liver fibrosis.
Collapse
|
20
|
Pu ZH, Liu J, Peng C, Luo M, Zhou QM, Xie XF, Chen MH, Xiong L. Nucleoside alkaloids with anti-platelet aggregation activity from the rhizomes of Ligusticum striatum. Nat Prod Res 2017; 33:1399-1405. [DOI: 10.1080/14786419.2017.1416382] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Affiliation(s)
- Zhong-Hui Pu
- State Key Laboratory Breeding Base of Systematic Research, Development and Utilization of Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- College of Life Science and Technology of Mianyang Normal University, MianYang, China
| | - Juan Liu
- State Key Laboratory Breeding Base of Systematic Research, Development and Utilization of Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Institute of Innovative Medicine Ingredients of Southwest Specialty Medicinal Materials, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Cheng Peng
- State Key Laboratory Breeding Base of Systematic Research, Development and Utilization of Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Institute of Innovative Medicine Ingredients of Southwest Specialty Medicinal Materials, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Min Luo
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Qin-Mei Zhou
- State Key Laboratory Breeding Base of Systematic Research, Development and Utilization of Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Institute of Innovative Medicine Ingredients of Southwest Specialty Medicinal Materials, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiao-Fang Xie
- State Key Laboratory Breeding Base of Systematic Research, Development and Utilization of Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Institute of Innovative Medicine Ingredients of Southwest Specialty Medicinal Materials, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Ming-Hua Chen
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Liang Xiong
- State Key Laboratory Breeding Base of Systematic Research, Development and Utilization of Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Institute of Innovative Medicine Ingredients of Southwest Specialty Medicinal Materials, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
21
|
Zhou Y, Ji Z, Yan W, Zhou Z, Li H, Xiao Y. Tetramethylpyrazine inhibits prostate cancer progression by downregulation of forkhead box M1. Oncol Rep 2017; 38:837-842. [PMID: 28677763 PMCID: PMC5561788 DOI: 10.3892/or.2017.5768] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Accepted: 06/19/2017] [Indexed: 12/13/2022] Open
Abstract
Tetramethylpyrazine (TMP) has exhibited various anticancer effects. However, its ability to inhibit proliferation, migration, and invasion of prostate cancer (PCa) PC-3 cells is still unclear. In the present study, different concentrations of TMP were co-incubated with PC-3 cells. The pcDNA-FOXM1 plasmid was transfected into cells before treatment with 500 µg/l TMP. The proliferative, migratory and invasive abilities of PC-3 cells were tested by MTT assay, wound healing assay and colony formation assay. Western blotting was used to investigate the expression of FOXM1. We found that, compared with the control, the proliferative, migratory and invasive abilities of PC-3 cells were decreased after incubation with different concentrations of TMP (P<0.01). The expression of FOXM1 was decreased in TMP-treated PC-3 cells (P<0.01). In addition, overexpression of FOXM1 reversed TMP-mediated inhibition of proliferation, migration and invasion of PC-3 cells. We also found that TMP inhibited PCa growth in vivo in a dose-dependent manner. These results suggest that TMP inhibits PC-3 cell proliferation, migration and invasion by downregulation of FOXM1.
Collapse
Affiliation(s)
- Yi Zhou
- Department of Urology, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100000, P.R. China
| | - Zhigang Ji
- Department of Urology, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100000, P.R. China
| | - Weigang Yan
- Department of Urology, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100000, P.R. China
| | - Zhien Zhou
- Department of Urology, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100000, P.R. China
| | - Hanzhong Li
- Department of Urology, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100000, P.R. China
| | - Yu Xiao
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100000, P.R. China
| |
Collapse
|
22
|
Wang Y, Guo G, Yang BR, Xin QQ, Liao QW, Lee SMY, Hu YJ, Chen KJ, Cong WH. Synergistic effects of Chuanxiong-Chishao herb-pair on promoting angiogenesis at network pharmacological and pharmacodynamic levels. Chin J Integr Med 2017; 23:654-662. [PMID: 28551771 DOI: 10.1007/s11655-017-2408-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Indexed: 11/28/2022]
Abstract
OBJECTIVE To investigate the synergistic effects of Chuanxiong-Chishao herb-pair (CCHP) on promoting angiogenesis in silico and in vivo. METHODS The mechanisms of action of an herb-pair, Chuanxiong-Chishao, were investigated using the network pharmacological and pharmacodynamic strategies involving computational drug target prediction and network analysis, and experimental validation. A set of network pharmacology methods were created to study the herbs in the context of targets and diseases networks, including prediction of target profiles and pharmacological actions of main active compounds in Chuanxiong and Chishao. Furthermore, the therapeutic effects and putative molecular mechanisms of Chuanxiong-Chishao actions were experimentally validated in a chemical-induced vascular insuffificiency model of transgenic zebrafifish in vivo. The mRNA expression of the predicted targets were further analyzed by real-time polymerase chain reaction (RT-PCR). RESULTS The computational prediction results found that the compounds in Chuanxiong have antithrombotic, antihypertensive, antiarrhythmic, and antiatherosclerotic activities, which were closely related to protecting against hypoxic-ischemic encephalopathy, ischemic stroke, myocardial infarction and heart failure. In addition, compounds in Chishao were found to participate in anti-inflflammatory effect and analgesics. Particularly, estrogen receptor α (ESRα) and hypoxia-inducible factor 1-α (HIF-1α) were the most important potential protein targets in the predicted results. In vivo experimental validation showed that post-treatment of tetramethylpyrazine hydrochloride (TMP•HCl) and paeoniflorin (PF) promoted the regeneration of new blood vessels in zebrafifish involving up-regulating ESRα mRNA expression. Co-treatment of TMP•HCl and PF could enhance the vessel sprouting in chemical-induced vascular insuffificiency zebrafifish at the optimal compatibility proportion of PF 10 μmol/L with TMP•HCl 1 μmol/L. CONCLUSIONS The network pharmacological strategies combining drug target prediction and network analysis identified some putative targets of CCHP. Moreover, the transgenic zebrafifish experiments demonstrated that the Chuanxiong-Chishao combination synergistically promoted angiogenic activity, probably involving ESRα signaling pathway.
Collapse
Affiliation(s)
- Yan Wang
- Clinical Medical College, Beijing University of Chinese Medicine, Beijing, 100029, China.,Laboratory of Cardiovascular Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, 100091, China.,State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR, 999078, China
| | - Gang Guo
- Department of Acupuncture and Moxibustion, Qilu Hospital Affiliated to Shandong University, Jinan, 250012, China
| | - Bin-Rui Yang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR, 999078, China
| | - Qi-Qi Xin
- Laboratory of Cardiovascular Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, 100091, China
| | - Qi-Wen Liao
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR, 999078, China
| | - Simon Ming-Yuen Lee
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR, 999078, China
| | - Yuan-Jia Hu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR, 999078, China
| | - Ke-Ji Chen
- Laboratory of Cardiovascular Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, 100091, China.
| | - Wei-Hong Cong
- Laboratory of Cardiovascular Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, 100091, China
| |
Collapse
|
23
|
Chinese Herbal Extractions for Relieving Radiation Induced Lung Injury: A Systematic Review and Meta-Analysis. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2017; 2017:2141645. [PMID: 28465702 PMCID: PMC5390604 DOI: 10.1155/2017/2141645] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Accepted: 03/09/2017] [Indexed: 11/17/2022]
Abstract
Background. Radiation induced lung injury (RILI) is one of the most common and severe side effects of thoracic radiotherapy. In this meta-analysis, the effects of Chinese herbal extractions (CHE) for preventing and treating RILI are evaluated. Methods. Randomized Controlled Trials (RCTs) from five databases were identified. Studies were evaluated and the relevant data were extracted by two authors independently. Differences were resolved by a third party. Meta-analysis was conducted using RevMan 5.0. Results. In total, 2734 participants receiving thoracic radiotherapy were included in 28 RCTs, and 16 CHE were evaluated. Meta-analysis showed that CHE intervention significantly reduced the incidence of acute radiation pneumonitis (RP) and radiation induced pulmonary fibrosis (RIPF). In CHE group, total effective rate and remission rate of RILI patients were significantly higher. Patient's quality of life (Qol) and clinical symptoms and signs were improved significantly. Inflammatory cytokines decreased, and thymus dependent lymphocytes subgroups were improved. Conclusion. CHE intervention may have clinical effectiveness for relieving RILI and related symptoms and signs and lead to improvement of Qol. However, more double-blind, multicenter, large-scale RCTs are needed to support this theory. Trial Registration. PROSPERO International prospective register of systematic reviews has registration number CRD42016043538.
Collapse
|
24
|
Park JS, Yi SW, Kim HJ, Kim SM, Park KH. Regulation of Cell Signaling Factors Using PLGA Nanoparticles Coated/Loaded with Genes and Proteins for Osteogenesis of Human Mesenchymal Stem Cells. ACS APPLIED MATERIALS & INTERFACES 2016; 8:30387-30397. [PMID: 27792311 DOI: 10.1021/acsami.6b08343] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Transfection of specific genes and transportation of proteins into cells have been a focus of stem cell differentiation research. However, it is not easy to regulate codelivery of a gene and a protein into cells. For codelivery into undifferentiated cells (human mesenchymal stem cells (hMSCs)), we used biodegradable carriers loaded with Runt-related transcription factor 2 (RUNX2) protein and coated with bone morphogenetic protein 2 (BMP2) plasmid DNA (pDNA) to induce osteogenesis. The released gene and protein were first localized in the cytosol of transfected hMSCs, and the gene then moved into the nucleus. The levels of internalized PLGA nanoparticles were tested using different doses and incubation durations. Then, transfection of BMP2 pDNA was confirmed by determining mRNA and protein levels and acquiring cell images. The same techniques were used to assess osteogenesis of hMSCs both in vitro and in vivo upon internalization of PLGA NPs carrying the BMP2 gene and RUNX2 protein. Detection of specific genes and proteins demonstrated that cells transfected with PLGA NPs carrying both the BMP2 gene and RUNX2 protein were highly differentiated compared with other samples. Histological and immunofluorescence analyses demonstrated that transfection of PLGA nanoparticles carrying both the BMP2 gene and RUNX2 protein dramatically enhanced osteogenesis of hMSCs.
Collapse
Affiliation(s)
- Ji Sun Park
- Department of Biomedical Science, College of Life Science, CHA University , 6F CHA Bio-complex, 689 Sampyeong-dong Bundang-gu, Seongnam-si, 463-400, Korea
| | - Se Won Yi
- Department of Biomedical Science, College of Life Science, CHA University , 6F CHA Bio-complex, 689 Sampyeong-dong Bundang-gu, Seongnam-si, 463-400, Korea
| | - Hye Jin Kim
- Department of Biomedical Science, College of Life Science, CHA University , 6F CHA Bio-complex, 689 Sampyeong-dong Bundang-gu, Seongnam-si, 463-400, Korea
| | - Seong Min Kim
- Department of Biomedical Science, College of Life Science, CHA University , 6F CHA Bio-complex, 689 Sampyeong-dong Bundang-gu, Seongnam-si, 463-400, Korea
| | - Keun-Hong Park
- Department of Biomedical Science, College of Life Science, CHA University , 6F CHA Bio-complex, 689 Sampyeong-dong Bundang-gu, Seongnam-si, 463-400, Korea
| |
Collapse
|
25
|
Luan Y, Liu J, Liu X, Xue X, Kong F, Sun C, Wang J, Liu L, Jia H. Tetramethypyrazine inhibits renal cell carcinoma cells through inhibition of NKG2D signaling pathways. Int J Oncol 2016; 49:1704-12. [DOI: 10.3892/ijo.2016.3670] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Accepted: 08/02/2016] [Indexed: 11/05/2022] Open
|