51
|
Lewoniewska S, Oscilowska I, Forlino A, Palka J. Understanding the Role of Estrogen Receptor Status in PRODH/POX-Dependent Apoptosis/Survival in Breast Cancer Cells. BIOLOGY 2021; 10:biology10121314. [PMID: 34943229 PMCID: PMC8698543 DOI: 10.3390/biology10121314] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 11/29/2021] [Accepted: 12/07/2021] [Indexed: 04/15/2023]
Abstract
It has been suggested that activation of estrogen receptor α (ER α) stimulates cell proliferation. In contrast, estrogen receptor β (ER β) has anti-proliferative and pro-apoptotic activity. Although the role of estrogens in estrogen receptor-positive breast cancer progression has been well established, the mechanism of their effect on apoptosis is not fully understood. It has been considered that ER status of breast cancer cells and estrogen availability might determine proline dehydrogenase/proline oxidase (PRODH/POX)-dependent apoptosis. PRODH/POX is a mitochondrial enzyme that converts proline into pyrroline-5-carboxylate (P5C). During this process, ATP (adenosine triphosphate) or ROS (reactive oxygen species) are produced, facilitating cell survival or death, respectively. However, the critical factor in driving PRODH/POX-dependent functions is proline availability. The amount of this amino acid is regulated at the level of prolidase (proline releasing enzyme), collagen biosynthesis (proline utilizing process), and glutamine, glutamate, α-ketoglutarate, and ornithine metabolism. Estrogens were found to upregulate prolidase activity and collagen biosynthesis. It seems that in estrogen receptor-positive breast cancer cells, prolidase supports proline for collagen biosynthesis, limiting its availability for PRODH/POX-dependent apoptosis. Moreover, lack of free proline (known to upregulate the transcriptional activity of hypoxia-inducible factor 1, HIF-1) contributes to downregulation of HIF-1-dependent pro-survival activity. The complex regulatory mechanism also involves PRODH/POX expression and activity. It is induced transcriptionally by p53 and post-transcriptionally by AMPK (AMP-activated protein kinase), which is regulated by ERs. The review also discusses the role of interconversion of proline/glutamate/ornithine in supporting proline to PRODH/POX-dependent functions. The data suggest that PRODH/POX-induced apoptosis is dependent on ER status in breast cancer cells.
Collapse
Affiliation(s)
- Sylwia Lewoniewska
- Department of Medicinal Chemistry, Medical University of Bialystok, Kilinskiego 1, 15-089 Bialystok, Poland;
| | - Ilona Oscilowska
- Department of Analysis and Bioanalysis of Medicines, Medical University of Bialystok, Kilinskiego 1, 15-089 Bialystok, Poland;
| | - Antonella Forlino
- Department of Molecular Medicine, University of Pavia, Viale Taramelli 3/B, 27100 Pavia, Italy;
| | - Jerzy Palka
- Department of Medicinal Chemistry, Medical University of Bialystok, Kilinskiego 1, 15-089 Bialystok, Poland;
- Correspondence: ; Tel.: +48-85-748-5706
| |
Collapse
|
52
|
Nurcahyanti ADR, Jap A, Lady J, Prismawan D, Sharopov F, Daoud R, Wink M, Sobeh M. Function of selected natural antidiabetic compounds with potential against cancer via modulation of the PI3K/AKT/mTOR cascade. Biomed Pharmacother 2021; 144:112138. [PMID: 34750026 DOI: 10.1016/j.biopha.2021.112138] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 08/19/2021] [Accepted: 08/31/2021] [Indexed: 12/12/2022] Open
Abstract
Diabetes mellitus (DM) is a metabolic disorder with growing global incidence, as 387 million people were diagnosed in 2014 with an expected projection of 642 million in 2040. Several complications are associated with DM including heart attack, stroke, kidney failure, blindness, and cancer. The latter is the second leading cause of death worldwide accounting for one in every six deaths, with liver, pancreas, and endometrium cancers are the most abundant among patients with diabetes. Phosphoinositide 3-kinase (PI3K)/protein kinase B (Akt)/mammalian target of rapamycin (mTOR) pathway plays a vital role in developing a wide array of pathological disorders, among them diabetes and cancer. Natural secondary metabolites that counteract the deleterious effects of reactive oxygen species (ROS) and modulate PI3K/Akt/mTOR pathway could be a promising approach in cancer therapy. Here, 717 medicinal plants with antidiabetic activities were highlighted along with 357 bioactive compounds responsible for the antidiabetic activity. Also, 43 individual plant compounds with potential antidiabetic activities against cancer via the modulation of PI3K/Akt/mTOR cascade were identified. Taken together, the available data give an insight of the potential of repurposing medicinal plants and/or the individual secondary metabolites with antidiabetic activities for cancer therapy.
Collapse
Affiliation(s)
- Agustina Dwi Retno Nurcahyanti
- Department of Pharmacy, School of Medicine and Health Sciences, Atma Jaya Catholic University of Indonesia, Pluit Raya 2, 14440 Jakarta, Indonesia.
| | - Adeline Jap
- Department of Pharmacy, School of Medicine and Health Sciences, Atma Jaya Catholic University of Indonesia, Pluit Raya 2, 14440 Jakarta, Indonesia
| | - Jullietta Lady
- Department of Pharmacy, School of Medicine and Health Sciences, Atma Jaya Catholic University of Indonesia, Pluit Raya 2, 14440 Jakarta, Indonesia
| | - Deka Prismawan
- Department of Pharmacy, School of Medicine and Health Sciences, Atma Jaya Catholic University of Indonesia, Pluit Raya 2, 14440 Jakarta, Indonesia
| | - Farukh Sharopov
- Chinese-Tajik Innovation Center for Natural Products, National Academy of Sciences of Tajikistan, Ayni str. 299/2, 734063, Dushanbe, Tajikistan
| | - Rachid Daoud
- African Genome Center, Mohammed VI Polytechnic University (UM6P), Lot 660, Hay Moulay Rachid, Ben Guerir 43150, Morocco
| | - Michael Wink
- Institute of Pharmacy and Molecular Biotechnology, Heidelberg University, Im Neuenheimer Feld 364, 69120 Heidelberg, Germany
| | - Mansour Sobeh
- AgroBiosciences Research, Mohammed VI Polytechnic University, Lot 660-Hay Moulay Rachid, 43150 Ben-Guerir, Morocco.
| |
Collapse
|
53
|
Xing Y, Lin Y, Zhang Y, Hu J, Liu J, Tian Y, Zhao J, Chen W, Han B. Novel cytoplasmic lncRNA IKBKBAS promotes lung adenocarcinoma metastasis by upregulating IKKβ and consequential activation of NF-κB signaling pathway. Cell Death Dis 2021; 12:1004. [PMID: 34702815 PMCID: PMC8548314 DOI: 10.1038/s41419-021-04304-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 10/01/2021] [Accepted: 10/11/2021] [Indexed: 02/08/2023]
Abstract
NF-κB signaling pathway is a critical link between inflammation and cancer. Emerging evidence suggested that long non-coding RNAs (lncRNAs) were involved in dysregulation of NF-κB. Herein, we reported a novel lncRNA IKBKBAS that activated NF-κB in lung adenocarcinoma (LUAD) by upregulating IKKβ, a key member of NF-κB signaling pathway, thereby promoting the metastasis of LUAD both in vitro and in vivo. The upregulated IKBKBAS functioned as a competing endogenous RNA (ceRNA) via competing with IKKβ mRNA for binding miR-4741, consequently leading to upregulation and activation of IKKβ, and ultimately activation of NF-κB. The abnormally elevated IKBKBAS in LUAD was mainly resulted from the extremely decrease of miR-512-5p that targeting IKBKBAS. Furthermore, we identified a positive feedback loop between NF-κB and IKBKBAS, in which NF-κB activation induced by overexpression of IKBKBAS could promote the transcription of IKBKBAS by binding the κB sites within IKBKBAS promoter. Our studies revealed that IKBKBAS was involved in the activation of NF-κB signaling by upregulating the expression of IKKβ, which made it serve as a potential novel target for therapies to LUAD.
Collapse
Affiliation(s)
- Yuanxin Xing
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
| | - Yani Lin
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
| | - Ying Zhang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
| | - Jing Hu
- Department of Pathology, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
| | - Junmei Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
| | - Yuanyuan Tian
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
| | - Jian Zhao
- Department of Thoracic Surgery, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China.
| | - Weiwen Chen
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China.
| | - Bo Han
- Department of Pathology, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Pathology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
| |
Collapse
|
54
|
Huang HC, Liao CC, Wang SH, Lee IJ, Lee TA, Hsu JM, Kuo CT, Wang J, Hsieh WC, Chang SJ, Chen SY, Tao MH, Lin YL, Lai YJ, Li CW. Hyperglycosylated spike of SARS-CoV-2 gamma variant induces breast cancer metastasis. Am J Cancer Res 2021; 11:4994-5005. [PMID: 34765306 PMCID: PMC8569360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Accepted: 07/10/2021] [Indexed: 06/13/2023] Open
Abstract
SARS-CoV-2 exploits the host cellular machinery for virus replication leading to the acute syndrome of coronavirus disease 2019 (COVID-19). Growing evidence suggests SARS-CoV-2 also exacerbates many chronic diseases, including cancers. As mutations on the spike protein (S) emerged as dominant variants that reduce vaccine efficacy, little is known about the relation between SARS-CoV-2 virus variants and cancers. Compared to the SARS-CoV-2 wild-type, the Gamma variant contains two additional NXT/S glycosylation motifs on the S protein. The hyperglycosylated S of Gamma variant is more stable, resulting in more significant epithelial-mesenchymal transition (EMT) potential. SARS-CoV-2 infection promoted NF-κB signaling activation and p65 nuclear translocation, inducing Snail expression. Pharmacologic inhibition of NF-κB activity by nature food compound, I3C suppressed viral replication and Gamma variant-mediated breast cancer metastasis, indicating that NF-κB inhibition can reduce chronic disease in COVID-19 patients. Our study revealed that the Gamma variant of SARS-CoV-2 activates NF-κB and, in turn, triggers the pro-survival function for cancer progression.
Collapse
Affiliation(s)
- Hsiang-Chi Huang
- Institute of Biomedical Sciences, Academia SinicaTaipei 115, Taiwan
| | - Chun-Che Liao
- Institute of Biomedical Sciences, Academia SinicaTaipei 115, Taiwan
| | - Shih-Han Wang
- Institute of Biomedical Sciences, Academia SinicaTaipei 115, Taiwan
| | - I-Jung Lee
- Institute of Biomedical Sciences, Academia SinicaTaipei 115, Taiwan
| | - Te-An Lee
- Institute of Biomedical Sciences, Academia SinicaTaipei 115, Taiwan
| | - Jung-Mao Hsu
- Graduate Institute of Biomedical Sciences and Research Center for Cancer Biology, China Medical UniversityTaichung 406040, Taiwan
| | - Chun-Tse Kuo
- Institute of Biomedical Sciences, Academia SinicaTaipei 115, Taiwan
| | - Jyun Wang
- Institute of Biomedical Sciences, Academia SinicaTaipei 115, Taiwan
| | - Wan-Chen Hsieh
- Institute of Biomedical Sciences, Academia SinicaTaipei 115, Taiwan
| | - Shing-Jyh Chang
- Department of Obstetrics and Gynecology, Hsinchu MacKay Memorial HospitalHsinchu 300, Taiwan
| | - Shih-Yu Chen
- Institute of Biomedical Sciences, Academia SinicaTaipei 115, Taiwan
| | - Mi-Hua Tao
- Institute of Biomedical Sciences, Academia SinicaTaipei 115, Taiwan
- Biomedical Translational Research Center, Academia SinicaTaipei 115, Taiwan
| | - Yi-Ling Lin
- Institute of Biomedical Sciences, Academia SinicaTaipei 115, Taiwan
- Biomedical Translational Research Center, Academia SinicaTaipei 115, Taiwan
| | - Yun-Ju Lai
- Institute of Biomedical Sciences, Academia SinicaTaipei 115, Taiwan
- Solomont School of Nursing, Zuckerberg College of Health Sciences, University of Massachusetts Lowell113 Wilder Street, Lowell, MA 01854, USA
| | - Chia-Wei Li
- Institute of Biomedical Sciences, Academia SinicaTaipei 115, Taiwan
| |
Collapse
|
55
|
Vageli DP, Doukas SG, Doukas PG, Judson BL. Bile reflux and hypopharyngeal cancer (Review). Oncol Rep 2021; 46:244. [PMID: 34558652 PMCID: PMC8485019 DOI: 10.3892/or.2021.8195] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 08/24/2021] [Indexed: 12/26/2022] Open
Abstract
Laryngopharyngeal reflux, a variant of gastroesophageal reflux disease, has been considered a risk factor in the development of hypopharyngeal cancer. Bile acids are frequently present in the gastroesophageal refluxate and their effect has been associated with inflammatory and neoplastic changes in the upper aerodigestive tract. Recent in vitro and in vivo studies have provided direct evidence of the role of acidic bile refluxate in hypopharyngeal carcinogenesis and documented the crucial role of NF-κB as a key mediator of early oncogenic molecular events in this process and also suggested a contribution of STAT3. Acidic bile can cause premalignant changes and invasive squamous cell cancer in the affected hypopharynx accompanied by DNA damage, elevated p53 expression and oncogenic mRNA and microRNA alterations, previously linked to head and neck cancer. Weakly acidic bile can also increase the risk for hypopharyngeal carcinogenesis by inducing DNA damage, exerting anti-apoptotic effects and causing precancerous lesions. The most important findings that strongly support bile reflux as an independent risk factor for hypopharyngeal cancer are presented in the current review and the underlying mechanisms are provided.
Collapse
Affiliation(s)
- Dimitra P Vageli
- The Yale Larynx Laboratory, Department of Surgery, Yale School of Medicine, New Haven, CT 06510, USA
| | - Sotirios G Doukas
- The Yale Larynx Laboratory, Department of Surgery, Yale School of Medicine, New Haven, CT 06510, USA
| | - Panagiotis G Doukas
- The Yale Larynx Laboratory, Department of Surgery, Yale School of Medicine, New Haven, CT 06510, USA
| | - Benjamin L Judson
- The Yale Larynx Laboratory, Department of Surgery, Yale School of Medicine, New Haven, CT 06510, USA
| |
Collapse
|
56
|
Cao RZ, Min L, Liu S, Tian RY, Jiang HY, Liu J, Shao LL, Cheng R, Zhu ST, Guo SL, Li P. Rictor Activates Cav 1 Through the Akt Signaling Pathway to Inhibit the Apoptosis of Gastric Cancer Cells. Front Oncol 2021; 11:641453. [PMID: 34540654 PMCID: PMC8442624 DOI: 10.3389/fonc.2021.641453] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 03/08/2021] [Indexed: 01/01/2023] Open
Abstract
Background Rapamycin-insensitive companion of mammalian target of rapamycin (Rictor) protein is a core subunit of mammalian target of rapamycin complex 2, and is associated with cancer progression. However, the biological function of Rictor in cancer, particularly its clinical relevance in gastric cancer (GC) remains largely unknown. Methods Rictor expression and its association with clinicopathologic characteristics in GC were analyzed by immunohistochemistry. Effect of Rictor and Caveolin-1 (Cav 1) on GC cells apoptosis was evaluated via overexpression experiment in vitro. Mechanisms of Rictor and Cav 1 in GC were explored through overexpression and knockdown, by immunofluorescence and western blot analyses. Results Rictor was upregulated in GC, and mainly located in the cytoplasm of cancer cells. Moreover, higher Rictor levels were associated with worse prognosis. Rictor could inhibit GC cell apoptosis and promote cell growth in vitro. The results of immunofluorescence revealed that Cav 1 localized in GC cell membrane but did not co-localize with Rictor. Further, Rictor regulated apoptosis-related proteins, long non-coding RNAs and also activated cellular signaling, thereby positively regulating Cav 1 expression. This effect was attenuated by the Akt inhibitor ly294002. Cav 1 did not significantly affect the ability of Rictor to inhibit tumor cell apoptosis. Conclusions Rictor is upregulated in GC and associated with worse prognosis. It inhibits tumor apoptosis and activates Cav 1 through the Akt signaling pathway to inhibit the apoptosis of GC cells. Rictor is, therefore, a promising prognostic biomarker and possible therapeutic target in GC patients.
Collapse
Affiliation(s)
- Rui-Zhen Cao
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, Beijing, China.,Department of Gastroenterology, Ordos Central Hospital, National Clinical Research Center for Digestive Disease-Ordos Subcenter, Ordos, China
| | - Li Min
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, Beijing, China
| | - Si Liu
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, Beijing, China
| | - Ru-Yue Tian
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, Beijing, China
| | - Hai-Yan Jiang
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, Beijing, China.,Department of Gastroenterology, Beijing University of Chinese Medicine Third Affiliated Hospital, Beijing, China
| | - Juan Liu
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, Beijing, China.,Department of Gastroenterology, Shanxi Province Cancer Hospital, Shanxi Medical University, Taiyuan, China
| | - Lin-Lin Shao
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, Beijing, China
| | - Rui Cheng
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, Beijing, China
| | - Sheng-Tao Zhu
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, Beijing, China
| | - Shui-Long Guo
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, Beijing, China
| | - Peng Li
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, Beijing, China
| |
Collapse
|
57
|
Mu Q, Najafi M. Resveratrol for targeting the tumor microenvironment and its interactions with cancer cells. Int Immunopharmacol 2021; 98:107895. [PMID: 34171623 DOI: 10.1016/j.intimp.2021.107895] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 06/09/2021] [Accepted: 06/13/2021] [Indexed: 12/17/2022]
Abstract
Tumor resistance to therapy modalities is one of the major challenges to the eradication of cancer cells and complete treatment. Tumor includes a wide range of cancer and non-cancer cells that play key roles in the proliferation of cancer cells and suppression of anti-tumor immunity. For overcoming tumor resistance to therapy, it is important to have in-depth knowledge relating to intercellular communications within the tumor microenvironment (TME). TME includes various types of immune cells such as CD4 + T lymphocytes, cytotoxic T lymphocytes (CTLs), natural killer (NK) cells, macrophages, and T regulatory cells (Tregs). Furthermore, some non-immune cells like cancer stem cells (CSCs), mesenchymal stem cells (MSCs), and cancer-associated fibroblasts (CAFs) are involved in the promotion of tumor growth. The interactions between these cells with cancer cells play a key role in tumor growth or inhibition. Resveratrol as a natural agent has shown the ability to modulate the immune system to potentiate anti-tumor immunity and also help to attenuate cancer cells and CSCs resistance. Thus, this review explains how resveratrol can modulate interactions within TME.
Collapse
Affiliation(s)
- Qi Mu
- College of Nursing, Inner Mongolia University for Nationalities, Tongliao 028000, China.
| | - Masoud Najafi
- Medical Technology Research Center, Institute of Health Technology, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| |
Collapse
|
58
|
Shao BY, Zhang SF, Li HD, Meng XM, Chen HY. Epigenetics and Inflammation in Diabetic Nephropathy. Front Physiol 2021; 12:649587. [PMID: 34025445 PMCID: PMC8131683 DOI: 10.3389/fphys.2021.649587] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Accepted: 04/12/2021] [Indexed: 12/11/2022] Open
Abstract
Diabetic nephropathy (DN) leads to high morbidity and disability. Inflammation plays a critical role in the pathogenesis of DN, which involves renal cells and immune cells, the microenvironment, as well as extrinsic factors, such as hyperglycemia, chemokines, cytokines, and growth factors. Epigenetic modifications usually regulate gene expression via DNA methylation, histone modification, and non-coding RNAs without altering the DNA sequence. During the past years, numerous studies have been published to reveal the mechanisms of epigenetic modifications that regulate inflammation in DN. This review aimed to summarize the latest evidence on the interplay of epigenetics and inflammation in DN, and highlight the potential targets for treatment and diagnosis of DN.
Collapse
Affiliation(s)
- Bao-Yi Shao
- Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Shao-Fei Zhang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China
| | - Hai-Di Li
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China
| | - Xiao-Ming Meng
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China
| | - Hai-Yong Chen
- School of Chinese Medicine, The University of Hong Kong, Hong Kong, China
- Department of Chinese Medicine, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| |
Collapse
|
59
|
BF 3-OEt 2 Catalyzed C3-Alkylation of Indole: Synthesis of Indolylsuccinimidesand Their Cytotoxicity Studies. Molecules 2021; 26:molecules26082202. [PMID: 33920456 PMCID: PMC8069703 DOI: 10.3390/molecules26082202] [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: 02/19/2021] [Revised: 03/27/2021] [Accepted: 04/02/2021] [Indexed: 11/17/2022] Open
Abstract
A simple and efficient BF3-OEt2 promoted C3-alkylation of indole has been developed to obtain3-indolylsuccinimidesfrom commercially available indoles and maleimides, with excellent yields under mild reaction conditions. Furthermore, anti-proliferative activity of these conjugates was evaluated against HT-29 (Colorectal), Hepg2 (Liver) and A549 (Lung) human cancer cell lines. One of the compounds, 3w, having N,N-Dimethylatedindolylsuccinimide is a potent congener amongst the series with IC50 value 0.02 µM and 0.8 µM against HT-29 and Hepg2 cell lines, respectively, and compound 3i was most active amongst the series with IC50 value 1.5 µM against A549 cells. Molecular docking study and mechanism of reaction have briefly beendiscussed. This method is better than previous reports in view of yield and substrate scope including electron deficient indoles.
Collapse
|
60
|
Jantan I, Haque MA, Arshad L, Harikrishnan H, Septama AW, Mohamed-Hussein ZA. Dietary polyphenols suppress chronic inflammation by modulation of multiple inflammation-associated cell signaling pathways. J Nutr Biochem 2021; 93:108634. [PMID: 33794330 DOI: 10.1016/j.jnutbio.2021.108634] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 01/20/2021] [Accepted: 03/10/2021] [Indexed: 02/06/2023]
Abstract
The high failure rate of the reductionist approach to discover effective and safe drugs to treat chronic inflammatory diseases has led scientists to seek alternative ways. Recently, targeting cell signaling pathways has been utilized as an innovative approach to discover drug leads from natural products. Cell signaling mechanisms have been identified playing key role in diverse diseases by inducing proliferation, cell survival and apoptosis. Phytochemicals are known to be able to modulate the cellular and molecular networks which are associated to chronic diseases including cancer-associated inflammation. In this review, the roles of dietary polyphenols (apigenin, kaempferol, quercetin, curcumin, genistein, isoliquiritigenin, resveratrol and gallic acid) in modulating multiple inflammation-associated cell signaling networks are deliberated. Scientific databases on suppressive effects of the polyphenols on chronic inflammation via modulation of the pathways especially in the recent five years are gathered and critically analyzed. The polyphenols are able to modulate several inflammation-associated cell signaling pathways, namely nuclear factor-kappa β, mitogen activated protein kinases, Wnt/β-catenin and phosphatidylinositol 3-kinase and protein kinase B via selective actions on various components of the networks. The suppressive effects of the polyphenols on the multiple cell signaling pathways reveal their potential use in prevention and treatment of chronic inflammatory disorders. Understanding the mechanistic effects involved in modulation of the signaling pathways by the polyphenols is necessary for lead identification and development of future functional foods for prevention and treatment of chronic inflammatory diseases.
Collapse
Affiliation(s)
- Ibrahim Jantan
- Institute of Systems Biology (INBIOSIS), Universiti Kebangsaan Malaysia, UKM Bangi, Selangor, Malaysia.
| | - Md Areeful Haque
- Department of Pharmacy, International Islamic University Chittagong, Chittagong, Bangladesh
| | - Laiba Arshad
- Department of Pharmacy, Forman Christian College (A Chartered University), Lahore, Pakistan
| | - Hemavathy Harikrishnan
- Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Abdi Wira Septama
- Research Center for Chemistry, Indonesian Institute of Sciences, Kawasan PUSPIPTEK Serpong, Tangerang Selatan, Banten, Indonesia
| | - Zeti-Azura Mohamed-Hussein
- Institute of Systems Biology (INBIOSIS), Universiti Kebangsaan Malaysia, UKM Bangi, Selangor, Malaysia; Department of Applied Physics, Faculty of Science & Technology, Universiti Kebangsaan Malaysia, UKM Bangi, Selangor
| |
Collapse
|
61
|
Circular RNAs as biomarkers and therapeutic targets in cancer. Semin Cancer Biol 2021; 83:242-252. [PMID: 33434640 DOI: 10.1016/j.semcancer.2020.12.026] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 11/25/2020] [Accepted: 12/30/2020] [Indexed: 02/06/2023]
Abstract
Circular RNAs (circRNAs) are a class of single-stranded closed non-coding RNA molecules (ncRNAs), which are formed as a result of reverse splicing of mRNAs. Despite their relative abundance, an interest in understanding their regulatory importance is rather recent. High stability, abundance and evolutionary conservation among species underline some of their important traits. CircRNAs perform a variety of cellular functions ranging from miRNA and proteins sponges to transcriptional modulation and splicing. Additionally, most circRNAs are expressed aberrantly in pathological conditions suggesting their possible exploitation as diagnostic biomarkers. Their covalent closed cyclic structure resulting in resistance to RNases further makes them suitable as cancer biomarkers. Studies involving human tumors have verified differences in the expression profiles of circRNAs, indicating a regulatory role in cancer pathogenesis and metastasis. As endogenous competitive RNA, circRNAs can regulate tumor proliferation and invasion. Further, some circRNAs located in the nucleus can regulate transcription of genes by binding to RNA polymerase II. In this review, we elaborate the characteristics, functions and mechanisms of action of circRNAs in cancer. We also discuss the possibility of using circRNAs as potential therapeutic targets and biomarkers for cancer.
Collapse
|
62
|
Laiakis EC, McCart EA, Deziel A, Rittase WB, Bouten RM, Jha J, Wilkins WL, Day RM, Fornace AJ. Effect of 3,3'-Diindolylmethane on Pulmonary Injury Following Thoracic Irradiation in CBA Mice. HEALTH PHYSICS 2020; 119:746-757. [PMID: 32384373 PMCID: PMC8579862 DOI: 10.1097/hp.0000000000001257] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
The molecule 3,3'-diindolylmethane (DIM) is small, a major bioactive metabolite of indole-3 carbinol (13C), and a phytochemical compound from cruciferous vegetables released upon exposure to the gut acid environment. DIM is a proposed anti-cancer agent and was previously demonstrated to prevent radiation damage in the bone marrow and the gastrointestinal tract. Here we investigated the effect of DIM on radiation-induced injury to the lung in a murine model through untargeted metabolomics and gene expression studies of select genes. CBA mice were exposed to thoracic irradiation (17.5 Gy). Mice were treated with vehicle or DIM (250 mg kg, subcutaneous injection) on days -1 pre-irradiation through +14 post-irradiation. DIM induced a significant improvement in survival by day 150 post-irradiation. Fibrosis-related gene expression and metabolomics were examined using lung tissue from days 15, 45, 60, 90, and 120 post-irradiation. Our qRT-PCR experiments showed that DIM treatment reduced radiation-induced late expression of collagen Iα and the cell cycle checkpoint proteins p21/waf1 (CDKN1A) and p16ink (CDKN2A). Metabolomic studies of lung tissue demonstrated a significant dampening of radiation-induced changes following DIM treatment. Metabolites associated with pro-inflammatory responses and increased oxidative stress, such as fatty acids, were suppressed by DIM treatment compared to irradiated samples. Together these data suggest that DIM reduces radiation-induced sequelae in the lung.
Collapse
Affiliation(s)
- Evagelia C. Laiakis
- Department of Oncology, Georgetown University, Washington, DC 20057, USA
- Department of Biochemistry and Molecular & Cellular Oncology, Georgetown University, Washington, DC 20057, USA
| | - Elizabeth A. McCart
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| | - Annabella Deziel
- Department of Oncology, Georgetown University, Washington, DC 20057, USA
| | - W. Bradley Rittase
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| | - Roxane M. Bouten
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| | - Jyoti Jha
- Current address: Rise Therapeutics, Rockville, MD 20850, USA
| | - W. Louis Wilkins
- Division of Comparative Pathology, the Armed Forces Radiobiology Research Institute/Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| | - Regina M. Day
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| | - Albert J. Fornace
- Department of Oncology, Georgetown University, Washington, DC 20057, USA
- Department of Biochemistry and Molecular & Cellular Oncology, Georgetown University, Washington, DC 20057, USA
| |
Collapse
|
63
|
Panada J, Klopava V, Kulahava T, Frolova N, Faletrov Y, Shkumatov V. New 3β-hydroxysteroid-indolamine conjugates: Design, synthesis and inhibition of C6 glioma cell proliferation. Steroids 2020; 164:108728. [PMID: 32931809 DOI: 10.1016/j.steroids.2020.108728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 08/21/2020] [Accepted: 09/07/2020] [Indexed: 10/23/2022]
Abstract
Four novel indole steroids based on dehydroepiandrosterone (IS-1), estrone (IS-2) and pregnenolone (IS-3) were obtained and studied for their ability to inhibit C6 glioma proliferation. A reduction in cell proliferation by 52 ± 13% was observed for IS-1 at 10 μM, whereas IS-3 and abiraterone acetate at 10 μM caused a 36 ± 8% decrease. Surprisingly, the cellular effects reported for abiraterone, namely, cytochrome P450 CYP17A1 inhibition and endoplasmic reticulum stress were not detected for IS-1. However, both abiraterone and IS-1 significantly increased glutathione levels. Docking studies predicted good affinity of IS-1 to liver X receptors and regulatory protein Keap1, which are proposed to be involved in the compounds' antiproliferative activity.
Collapse
Affiliation(s)
- Jan Panada
- Research Institute for Physical Chemical Problems of the Belarusian State University, Minsk, Belarus; Chemistry Faculty of Belarusian State University, Minsk, Belarus
| | - Valeriya Klopava
- Department of Biophysics, Physics Faculty of Belarusian State University, Minsk, Belarus
| | - Tatsiana Kulahava
- Department of Biophysics, Physics Faculty of Belarusian State University, Minsk, Belarus; Institute for Nuclear Problems of the Belarusian State University, Minsk, Belarus
| | - Nina Frolova
- Research Institute for Physical Chemical Problems of the Belarusian State University, Minsk, Belarus
| | - Yaroslav Faletrov
- Research Institute for Physical Chemical Problems of the Belarusian State University, Minsk, Belarus; Chemistry Faculty of Belarusian State University, Minsk, Belarus
| | - Vladimir Shkumatov
- Research Institute for Physical Chemical Problems of the Belarusian State University, Minsk, Belarus; Chemistry Faculty of Belarusian State University, Minsk, Belarus.
| |
Collapse
|
64
|
Wang X, Zhao Y, Yu M, Xu Y. PTEN/Akt Signaling-Mediated Activation of the Mitochondrial Pathway Contributes to the 3,3'-Diindolylmethane-Mediated Antitumor Effect in Malignant Melanoma Cells. J Med Food 2020; 23:1248-1258. [PMID: 33237846 DOI: 10.1089/jmf.2020.4704] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
3,3'-diindolylmethane (DIM) has an anticancer activity, but the role DIM plays on malignant melanoma cells and its specific mechanism is unclear. We studied the biological effects of DIM on malignant melanoma cells and the related mechanism and the results showed that DIM significantly suppressed cell proliferation and induced apoptosis in malignant melanoma cells. In addition, the expression levels of phosphatase and tensin homolog deleted on chromosome ten (PTEN), Bax, Bid, cleaved caspase-3, and cleaved caspase-9 were increased after DIM treatment. In A2058 PTENmut cells, DIM-mediated inhibition of proliferation and DIM-induced apoptosis were attenuated. Additionally, the overexpression and knockdown of PTEN could regulate such effects of DIM in malignant melanoma cells. Furthermore, DIM exerted growth-inhibiting and apoptosis-inducing effects in vivo. This study demonstrated that DIM has antitumor effect in human malignant melanoma cells through the mitochondrial apoptotic pathway activated by PTEN/Akt signaling.
Collapse
Affiliation(s)
- Xiaoli Wang
- Department of Plastic Surgery and Renmin Hospital of Wuhan University, Wuhan, China
| | - Yueqiang Zhao
- Department of Plastic Surgery and Renmin Hospital of Wuhan University, Wuhan, China
| | - Mosheng Yu
- Department of Plastic Surgery and Renmin Hospital of Wuhan University, Wuhan, China
| | - Yu Xu
- Department of Otolaryngology, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
65
|
Tran KB, Buchanan CM, Shepherd PR. Evolution of Molecular Targets in Melanoma Treatment. Curr Pharm Des 2020; 26:396-414. [PMID: 32000640 DOI: 10.2174/1381612826666200130091318] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2019] [Accepted: 11/21/2019] [Indexed: 12/12/2022]
Abstract
Melanoma is the deadliest type of skin cancers, accounting for more than 80% of skin cancer mortality. Although melanoma was known very early in the history of medicine, treatment for this disease had remained largely the same until very recently. Previous treatment options, including removal surgery and systemic chemotherapy, offered little benefit in extending the survival of melanoma patients. However, the last decade has seen breakthroughs in melanoma treatment, which all emerged following new insight into the oncogenic signaling of melanoma. This paper reviewed the evolution of drug targets for melanoma treatment based on the emergence of novel findings in the molecular signaling of melanoma. One of the findings that are most influential in melanoma treatment is that more than 50% of melanoma tumors contain BRAF mutations. This is fundamental for the development of BRAF inhibitors, which is the first group of drugs that significantly improves the overall survival of melanoma patients compared to the traditional chemotherapeutic dacarbazine. More recently, findings of the role of immune checkpoint molecules such as CTLA-4 and PD1/PD-L1 in melanoma biology have led to the development of a new therapeutic category: immune checkpoint inhibitors, which, for the first time in the history of cancer treatment, produced a durable response in a subset of melanoma patients. However, as this paper discussed next, there is still an unmet need for melanoma treatment. A significant population of patients did not respond to either BRAF inhibitors or immune checkpoint inhibitors. Of those patients who gained an initial response from those therapies, a remarkable percentage would develop drug resistance even when MEK inhibitors were added to the treatment. Finally, this paper discusses some possible targets for melanoma treatment.
Collapse
Affiliation(s)
- Khanh B Tran
- Department of Molecular Medicine and Pathology, University of Auckland, New Zealand.,Maurice Wilkins Centre for Molecular Biodiscovery, Auckland, New Zealand
| | - Christina M Buchanan
- Department of Molecular Medicine and Pathology, University of Auckland, New Zealand.,Maurice Wilkins Centre for Molecular Biodiscovery, Auckland, New Zealand
| | - Peter R Shepherd
- Department of Molecular Medicine and Pathology, University of Auckland, New Zealand.,Maurice Wilkins Centre for Molecular Biodiscovery, Auckland, New Zealand.,Auckland Cancer Society Research Centre, University of Auckland, New Zealand
| |
Collapse
|
66
|
Christofi M, Le Sommer S, Mölzer C, Klaska IP, Kuffova L, Forrester JV. Low-dose 2-deoxy glucose stabilises tolerogenic dendritic cells and generates potent in vivo immunosuppressive effects. Cell Mol Life Sci 2020; 78:2857-2876. [PMID: 33074350 PMCID: PMC8004500 DOI: 10.1007/s00018-020-03672-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 09/04/2020] [Accepted: 10/05/2020] [Indexed: 01/17/2023]
Abstract
Cell therapies for autoimmune diseases using tolerogenic dendritic cells (tolDC) have been promisingly explored. A major stumbling block has been generating stable tolDC, with low risk of converting to mature immunogenic DC (mDC), exacerbating disease. mDC induction involves a metabolic shift to lactate production from oxidative phosphorylation (OXPHOS) and β-oxidation, the homeostatic energy source for resting DC. Inhibition of glycolysis through the administration of 2-deoxy glucose (2-DG) has been shown to prevent autoimmune disease experimentally but is not clinically feasible. We show here that treatment of mouse bone marrow-derived tolDC ex vivo with low-dose 2-DG (2.5 mM) (2-DGtolDC) induces a stable tolerogenic phenotype demonstrated by their failure to engage lactate production when challenged with mycobacterial antigen (Mtb). ~ 15% of 2-DGtolDC express low levels of MHC class II and 30% express CD86, while they are negative for CD40. 2-DGtolDC also express increased immune checkpoint molecules PDL-1 and SIRP-1α. Antigen-specific T cell proliferation is reduced in response to 2-DGtolDC in vitro. Mtb-stimulated 2-DGtolDC do not engage aerobic glycolysis but respond to challenge via increased OXPHOS. They also have decreased levels of p65 phosphorylation, with increased phosphorylation of the non-canonical p100 pathway. A stable tolDC phenotype is associated with sustained SIRP-1α phosphorylation and p85-AKT and PI3K signalling inhibition. Further, 2-DGtolDC preferentially secrete IL-10 rather than IL-12 upon Mtb-stimulation. Importantly, a single subcutaneous administration of 2-DGtolDC prevented experimental autoimmune uveoretinitis (EAU) in vivo. Inhibiting glycolysis of autologous tolDC prior to transfer may be a useful approach to providing stable tolDC therapy for autoimmune/immune-mediated diseases.
Collapse
Affiliation(s)
- M Christofi
- Institute of Medical Sciences, School of Medicine, Medical Sciences and Nutrition, Foresterhill, University of Aberdeen, Aberdeen, AB25 2ZD, Scotland, UK
| | - S Le Sommer
- Institute of Medical Sciences, School of Medicine, Medical Sciences and Nutrition, Foresterhill, University of Aberdeen, Aberdeen, AB25 2ZD, Scotland, UK
| | - C Mölzer
- Institute of Medical Sciences, School of Medicine, Medical Sciences and Nutrition, Foresterhill, University of Aberdeen, Aberdeen, AB25 2ZD, Scotland, UK.
| | - I P Klaska
- Institute of Medical Sciences, School of Medicine, Medical Sciences and Nutrition, Foresterhill, University of Aberdeen, Aberdeen, AB25 2ZD, Scotland, UK
| | - L Kuffova
- Institute of Medical Sciences, School of Medicine, Medical Sciences and Nutrition, Foresterhill, University of Aberdeen, Aberdeen, AB25 2ZD, Scotland, UK.,Eye Clinic, Aberdeen Royal Infirmary, Aberdeen, Scotland, UK
| | - J V Forrester
- Institute of Medical Sciences, School of Medicine, Medical Sciences and Nutrition, Foresterhill, University of Aberdeen, Aberdeen, AB25 2ZD, Scotland, UK. .,Ocular Immunology Program, Centre for Ophthalmology and Visual Science, The University of Western Australia, Perth, WA, Australia. .,Centre for Experimental Immunology, Lions Eye Institute, Nedlands, WA, Australia.
| |
Collapse
|
67
|
Biersack B. 3,3'-Diindolylmethane and its derivatives: nature-inspired strategies tackling drug resistant tumors by regulation of signal transduction, transcription factors and microRNAs. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2020; 3:867-878. [PMID: 35582221 PMCID: PMC8992569 DOI: 10.20517/cdr.2020.53] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 08/31/2020] [Accepted: 09/22/2020] [Indexed: 11/14/2022]
Abstract
Indoles of cruciferous vegetables are promising anti-tumor agents. Studies with indole-3-carbinol and its dimeric product, 3,3'-diindolylmethane (DIM), suggest that these compounds have the ability to deregulate multiple cellular signaling pathways that are essential for tumor growth and spread. These natural compounds are also effective modulators of transcription factors and non-coding RNAs. These effects explain their ability to inhibit tumor spread and to overcome drug resistance. In this work, pertinent literature on the effects of DIM and its synthetic derivatives on resistant tumors and resistance mechanisms in tumors is highlighted.
Collapse
Affiliation(s)
- Bernhard Biersack
- Organic Chemistry 1, University of Bayreuth, Bayreuth 95440, Germany
| |
Collapse
|
68
|
Wu D, Li S, Liu X, Xu J, Jiang A, Zhang Y, Liu Z, Wang J, Zhou E, Wei Z, Yang Z, Guo C. Alpinetin prevents inflammatory responses in OVA-induced allergic asthma through modulating PI3K/AKT/NF-κB and HO-1 signaling pathways in mice. Int Immunopharmacol 2020; 89:107073. [PMID: 33039967 DOI: 10.1016/j.intimp.2020.107073] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 09/29/2020] [Accepted: 10/01/2020] [Indexed: 12/11/2022]
Abstract
Allergic asthma is the most common type of asthma which characterized by inflammatory responses of the airways. Alpinetin, a flavonoid compound derived from the ginger family of medicinal herbs, possesses various biological properties including anti-inflammatory, anti-oxidant and other medical effects. In this study, we aimed to evaluate the effects of alpinetin on OVA-induced allergic asthma, and further to examine its molecular mechanisms underlying these processes in vivo and in vitro. Mice were sensitized and challenged with OVA to build allergic asthma model in vivo. Bronchoalveolar lavage fluid (BALF) was collected for inflammatory cells analysis and lung tissues were examined for histopathological examination. The levels of IL-5, IL-13, IL-4, IgE, TNF-α, IL-6 and IL-1β were determined by the respective ELISA kits. The PI3K/AKT/NF-κB and HO-1 signaling pathways were examined by western blot analysis. The results showed that alpinetin significantly ameliorated OVA-induced pathologic changes of lungs, such as decreasing massive inflammatory cell infiltration and mucus hypersecretion, and reduced the number of inflammatory cells in BALF. Alpinetin also decreased the OVA-induced levels of IL-4, IL-5, IL-13 and IgE. Furthermore, alpinetin inhibited OVA-induced phosphorylation of p65, IκB, PI3K and AKT, and the activity of HO-1 in vivo. More importantly, these anti-inflammatory effects and molecular mechanisms of alpinetin has also been confirmed in LPS-stimulated RAW 264.7 macrophages in vitro. In conclusion, above results indicate that alpinetin exhibites a potent anti-inflammatory activity in allergic asthma through modulating PI3K/AKT/NF-κB and HO-1 signaling pathways, which would be used as a promising therapy agent for allergic asthma.
Collapse
Affiliation(s)
- Di Wu
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun 130062, Jilin Province, PR China
| | - Shuangqiu Li
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun 130062, Jilin Province, PR China
| | - Xiao Liu
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun 130062, Jilin Province, PR China
| | - Jingnan Xu
- College of Life Sciences and Engineering, Foshan University, Foshan 528225, Guangdong Province, PR China
| | - Aimin Jiang
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun 130062, Jilin Province, PR China
| | - Yong Zhang
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun 130062, Jilin Province, PR China
| | - Ziyi Liu
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun 130062, Jilin Province, PR China
| | - Jingjing Wang
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun 130062, Jilin Province, PR China
| | - Ershun Zhou
- College of Life Sciences and Engineering, Foshan University, Foshan 528225, Guangdong Province, PR China
| | - Zhengkai Wei
- College of Life Sciences and Engineering, Foshan University, Foshan 528225, Guangdong Province, PR China
| | - Zhengtao Yang
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun 130062, Jilin Province, PR China; College of Life Sciences and Engineering, Foshan University, Foshan 528225, Guangdong Province, PR China.
| | - Changmin Guo
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun 130062, Jilin Province, PR China.
| |
Collapse
|
69
|
Esteve M. Mechanisms Underlying Biological Effects of Cruciferous Glucosinolate-Derived Isothiocyanates/Indoles: A Focus on Metabolic Syndrome. Front Nutr 2020; 7:111. [PMID: 32984393 PMCID: PMC7492599 DOI: 10.3389/fnut.2020.00111] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Accepted: 06/16/2020] [Indexed: 12/13/2022] Open
Abstract
An inverse correlation between vegetable consumption and the incidence of cancer has long been described. This protective effect is stronger when cruciferous vegetables are specifically consumed. The beneficial properties of vegetables are attributed to their bioactive components like fiber, antioxidants vitamins, antioxidants, minerals, and phenolic compounds. Cruciferous vegetables contain all these molecules; however, what makes them different are their sulfurous components, called glucosinolates, responsible for their special smell and taste. Glucosinolates are inactive biologically in the organism but are hydrolyzed by the enzyme myrosinase released as a result of chewing, leading to the formation of active derivatives such as isothiocyanates and indoles. A considerable number of in vitro and in vivo studies have reported that isothiocyanates and indoles elicit chemopreventive potency through multiple mechanisms that include modulation of phases I and II detoxification pathway enzymes, regulation of cell cycle arrest, and control of cell growth, induction of apoptosis, antioxidant activity, anti-angiogenic effects, and epigenetic regulation. Nuclear erythroid 2-related factor 2 (Nrf2) and Nuclear factor-κB (NF-κB) are key and central regulators in all these processes with a main role in oxidative stress and inflammation control. It has been described that isothiocyanates and indoles regulate their activity directly and indirectly. Today, the metabolic syndrome (central obesity, insulin resistance, hyperlipidemia, and hypertension) is responsible for a majority of deaths worldwide. All components of metabolic syndrome are characterized by chronic inflammation with deregulation of the PI3K/AKT/mTOR, MAPK/EKR/JNK, Nrf2, and NF-κB signaling pathways. The effects of GLSs derivatives controlling these pathways have been widely described in relation to cancer. Changes in food consumption patterns observed in the last decades to higher consumption of ultra-processed foods, with elevation in simple sugar and saturated fat contents and lower consumption of vegetables and fruits have been directly correlated with metabolic syndrome prevalence. In this review, it is summarized the knowledge regarding the mechanisms by which cruciferous glucosinolate derivatives (isothiocyanates and indoles) directly and indirectly regulate these pathways. However, the review places a special focus on the knowledge of the effects of glucosinolates derivatives in metabolic syndrome, since this has not been reviewed before.
Collapse
Affiliation(s)
- Montserrat Esteve
- Department of Biochemistry and Molecular Biomedicine, University of Barcelona, Barcelona, Spain
- Biomedical Research Centre in Physiopathology of Obesity and Nutrition (CIBERobn), Institute of Health Carlos III, Madrid, Spain
| |
Collapse
|
70
|
Najafi M, Mortezaee K, Rahimifard M, Farhood B, Haghi-Aminjan H. The role of curcumin/curcuminoids during gastric cancer chemotherapy: A systematic review of non-clinical study. Life Sci 2020; 257:118051. [DOI: 10.1016/j.lfs.2020.118051] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 07/01/2020] [Accepted: 07/01/2020] [Indexed: 02/07/2023]
|
71
|
Apaza T L, Antognoni F, Potente G, Rumbero Sánchez Á. Triterpenoids isolated from Jatropha macrantha (Müll. Arg.) inhibit the NF-κB and HIF-1α pathways in tumour cells. Nat Prod Res 2020; 35:5843-5847. [PMID: 32691666 DOI: 10.1080/14786419.2020.1795851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Activity-guided fractionations of Jatropha macrantha Müll. Arg. led to the isolation of pomolic acid (1) and euscaphic acid (2). The potential for inhibition against NF-κB and HIF-1α production of these two compounds was tested in different tumour cell lines. Compounds 1 and 2 showed an inhibitory activity of HIF-1α in the SK-MEL-28 (IC50=3.01 ± 0.02 μM and 3.78 ± 0.02 μM), A549 (IC50=9.97 ± 0.01 μM and 10.25 ± 0.01 μM) and U-373 MG (IC50=6.34 ± 0.02 μM and 8.85 ± 0.02 μM) cell lines. In addition, compounds 1 and 2 showed an inhibitory activity on NF-κB in SK-MEL-28 (IC50=1.05 ± 0.02 μM and 2.71 ± 0.01 μM), A549 (IC50=3.63 ± 0.01 μM and 3.73 ± 0.02 μM) and U-373 MG (IC50=2.55 ± 0.02 μM and 3.39 ± 0.01 μM) cell lines. This is the first report that isolates these compounds from J. macrantha and tests their antitumor potential.
Collapse
Affiliation(s)
- Luis Apaza T
- Department of Organic Chemistry, Faculty of Sciences, University Autónoma of Madrid, Madrid, Spain.,Department of Pharmacology, Pharmacognosy and Botany, Faculty of Pharmacy, University Complutense of Madrid, Madrid, Spain
| | - Fabiana Antognoni
- Department for Life Quality Studies, University of Bologna, Rimini, Italy
| | - Giulia Potente
- Department for Life Quality Studies, University of Bologna, Rimini, Italy
| | - Ángel Rumbero Sánchez
- Department of Organic Chemistry, Faculty of Sciences, University Autónoma of Madrid, Madrid, Spain
| |
Collapse
|
72
|
Ciebiera M, Ali M, Prince L, Jackson-Bey T, Atabiekov I, Zgliczyński S, Al-Hendy A. The Evolving Role of Natural Compounds in the Medical Treatment of Uterine Fibroids. J Clin Med 2020; 9:E1479. [PMID: 32423112 PMCID: PMC7290481 DOI: 10.3390/jcm9051479] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 05/11/2020] [Accepted: 05/11/2020] [Indexed: 12/15/2022] Open
Abstract
Uterine fibroids (UFs) remain a significant health issue for many women, with a disproportionate impact on women of color, likely due to both genetic and environmental factors. The prevalence of UFs is estimated to be approximately 70% depending on population. UF-derived clinical symptoms include pelvic pain, excessive uterine bleeding, gastrointestinal and voiding problems, as well as impaired fertility. Nowadays numerous methods of UF treatment are available-from conservative treatment to invasive surgeries. Selecting an appropriate treatment option should be individualized and adjusted to the patient's expectations as much as possible. So far, the mainstay of treatment is surgery, but their negative impact of future fertility is clear. On the other hand, emerging new pharmaceutical options have significant adverse effects like liver function impairment, hot flashes, bone density loss, endometrial changes, and inability to attempt conception during treatment. Several natural compounds are found to help treat UFs and relieve their symptoms. In this review we summarize all the current available data about natural compounds that may be beneficial for patients with UFs, especially those who want to preserve their future fertility or have treatment while actively pursuing conception. Vitamin D, epigallocatechin gallate, berberine, curcumin, and others are being used as alternative UF treatments. Moreover, we propose the concept of using combined therapies of natural compounds on their own or combined with hormonal agents to manage UFs. There is a strong need for more human clinical trials involving these compounds before promoting widespread usage.
Collapse
Affiliation(s)
- Michał Ciebiera
- Second Department of Obstetrics and Gynecology, The Center of Postgraduate Medical Education, 01-809 Warsaw, Poland;
- Department of Surgery, University of Illinois at Chicago, Chicago, IL 60612, USA;
| | - Mohamed Ali
- Department of Surgery, University of Illinois at Chicago, Chicago, IL 60612, USA;
- Clinical Pharmacy Department, Faculty of Pharmacy, Ain Shams University, 11566 Cairo, Egypt
| | - Lillian Prince
- School of Public Health, University of Illinois at Chicago, Chicago, IL 60612, USA;
| | - Tia Jackson-Bey
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, University of Illinois at Chicago, Chicago, IL 60612, USA;
| | - Ihor Atabiekov
- Moscow Region Cancer Center, Balashikha 143900, Russian;
| | - Stanisław Zgliczyński
- Department of Internal Diseases and Endocrinology, Central Teaching Clinical Hospital, Medical University of Warsaw, 02-097 Warsaw, Poland;
| | - Ayman Al-Hendy
- Department of Surgery, University of Illinois at Chicago, Chicago, IL 60612, USA;
| |
Collapse
|
73
|
Mousa DS, El-Far AH, Saddiq AA, Sudha T, Mousa SA. Nanoformulated Bioactive Compounds Derived from Different Natural Products Combat Pancreatic Cancer Cell Proliferation. Int J Nanomedicine 2020; 15:2259-2268. [PMID: 32280218 PMCID: PMC7127850 DOI: 10.2147/ijn.s238256] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Accepted: 02/26/2020] [Indexed: 12/12/2022] Open
Abstract
Purpose This study was designed to determine the potential effect of nanoencapsulated bioactive compounds from different natural sources on human pancreatic cancer. Background Pancreatic cancer carries the highest fatality rate among all human cancers because of its high metastatic potential and late presentation at the time of diagnosis. Hence there is a need for improved methods to prevent and treat it. Natural products, such as 3, 3′-diindolylmethane (DIM) and ellagic acid (EA) demonstrated anticancer efficacy against various cancer types. However, DIM is insoluble. Hence, using nanotechnology to encapsulate these compounds in combination with EA might improve their physical and chemical properties and their delivery to the cancer cells. Methods Human pancreatic cancer cells, namely SUIT2-luciferase transfected, were used to examine the effects of DIM or EA and their nanoformulation in poly(lactic-co-glycolic acid) (PLGA) and poly(ethylene glycol) (PEG) [PLGA-PEG] nanoparticles (NPs) on SUIT2-luciferase cell viability/proliferation over 24 hrs. Additionally, effects on tumor weight and angiogenesis were determined using the chick chorioallantoic membrane (CAM) tumor implant model. Results Both DIM and EA PLGA-PEG NPs resulted in rapid suppression of pancreatic cancer cell viability/proliferation within 24 hrs (P < 0.01), while the non-encapsulated DIM and EA did not show any significant effect on SUIT2 cancer cell viability or cell proliferation (MTT assay). In the CAM pancreatic cancer cell (SUIT2) implant model, results showed a greater suppression of tumor weight (P < 0.01), tumor cell viability, and tumor angiogenesis (P < 0.01) for DIM NPs and EA NPs and their combinations versus DIM or EA alone. Conclusion Nanoformulation of DIM and EA resulted in a more effective suppression of pancreatic cancer cell viability, pancreatic tumor weight, implanted cancer cell viability, and tumor angiogenesis as compared with these bioactive compounds alone.
Collapse
Affiliation(s)
| | - Ali H El-Far
- Department of Biochemistry, Faculty of Veterinary Medicine, Damanhour University, Damanhour 22511, Egypt
| | - Amna A Saddiq
- Faculty of Science, Department of Biology, University of Jeddah, Jeddah 21589, Saudi Arabia
| | - Thangirala Sudha
- Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, Rensselaer, NY 12144, USA
| | - Shaker A Mousa
- Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, Rensselaer, NY 12144, USA
| |
Collapse
|
74
|
Li X, Shang D, Shen H, Song J, Hao G, Tian Y. ZSCAN16 promotes proliferation, migration and invasion of bladder cancer via regulating NF-kB, AKT, mTOR, P38 and other genes. Biomed Pharmacother 2020; 126:110066. [PMID: 32172065 DOI: 10.1016/j.biopha.2020.110066] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2019] [Revised: 01/16/2020] [Accepted: 01/23/2020] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND As one of the most common genitourinary malignancies worldwide, bladder cancer affects about 3.4 million people globally, with 430,000 new cases a year since 2015. Despite the advances in bladder cancer diagnosis and therapy, there has been little progress in the patients' overall survival in nearly 30 years. Therefore, investigating novel molecular therapeutic targets is required to gain insight into the tumorigenesis of bladder cancer, which ultimately may be used to develop more effective therapeutic strategies. METHODS Herein, we used gene knockdown in vitro and in vivo to unveil the unknown roles of ZSCAN16 in bladder cancer. Afterward, to decipher the unknown regulatory role of ZSCAN16 in tumor progression, we verified that a bunch of genes including NF-κB, AKT, mTOR, and P38 were the key downstream regulators of ZSCAN16 by western blot and rescue experiments. RESULTS We found high expression of ZSCAN16 transcripts in bladder cancer cells and tumor samples from the TCGA database and tissue microarray bank, demonstrated in correlation with poor prognosis for bladder cancer patients. The in vitro experiments indicated that the silencing of ZSCAN16 by shRNA lentivirus promoted apoptosis and inhibited proliferation, colony formation, as well as migration and invasion in T24 cells. By investigating the signaling pathways, we proved ZSCAN16 play a novel role as oncogenic gene in bladder cancer by regulating NF-κB, AKT, mTOR, P38 and other genes. Furthermore, the in vivo experiments identified that ZSCAN16 knockdown retarded the tumor growth in nude mice. CONCLUSIONS In summary, these findings revealed that ZSCAN16 is a potential novel oncogene in the development and progression of bladder cancer. This study will shed light on developing novel therapeutic targets in the future treatment of bladder cancer.
Collapse
Affiliation(s)
- Xuanhao Li
- Department of Urology, Beijing Friendship Hospital, No. 95 Yong'an Road, Xicheng District, Beijing, 100050, PR China.
| | - Donghao Shang
- Department of Urology, Beijing Friendship Hospital, No. 95 Yong'an Road, Xicheng District, Beijing, 100050, PR China.
| | - Hongliang Shen
- Department of Urology, Beijing Friendship Hospital, No. 95 Yong'an Road, Xicheng District, Beijing, 100050, PR China.
| | - Jian Song
- Department of Urology, Beijing Friendship Hospital, No. 95 Yong'an Road, Xicheng District, Beijing, 100050, PR China.
| | - Gangyue Hao
- Department of Urology, Beijing Friendship Hospital, No. 95 Yong'an Road, Xicheng District, Beijing, 100050, PR China.
| | - Ye Tian
- Department of Urology, Beijing Friendship Hospital, No. 95 Yong'an Road, Xicheng District, Beijing, 100050, PR China.
| |
Collapse
|
75
|
Li W, Du Q, Li X, Zheng X, Lv F, Xi X, Huang G, Yang J, Liu S. Eriodictyol Inhibits Proliferation, Metastasis and Induces Apoptosis of Glioma Cells via PI3K/Akt/NF-κB Signaling Pathway. Front Pharmacol 2020; 11:114. [PMID: 32158391 PMCID: PMC7052300 DOI: 10.3389/fphar.2020.00114] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Accepted: 01/28/2020] [Indexed: 11/13/2022] Open
Abstract
Glioma is the most common type of malignant brain tumor. Due to its highly aggressive and metastatic features, glioma is associated with poor prognosis and a lack of effective treatments. Eriodictyol, a natural flavonoid compound, has been reported to possess anti-inflammatory and antioxidant effects. However, the anti-tumor effects of eriodictyol and the underlying mechanisms have rarely been reported. In this study, we found that eriodictyol has anti-tumor activity in lung, colon, breast, pancreas, and liver cancer, and most significantly in glioma cell lines. Eriodictyol dose- and time-dependently suppresses cell proliferation, migration, and invasion in U87MG and CHG-5 glioma cells. In addition, eriodictyol induces apoptosis in U87MG and CHG-5 cells, as evaluated by flow cytometry, immunofluorescence, and Western blot. Furthermore, eriodictyol downregulates the phosphoinositide 3-kinase (PI3K)/Akt/NF-κB signaling pathway in a concentration-dependent manner. Moreover, the effects of eriodictyol on the apoptosis of glioma cells are enhanced by LY294002 (a PI3K inhibitor) and reversed by 740 Y-P (a PI3K agonist). In a mouse xenograft model, eriodictyol not only dramatically suppressed tumor growth but also induced apoptosis in tumor cells. In summary, our data illustrate that eriodictyol effectively inhibits proliferation and metastasis and induces apoptosis of glioma cell lines, which might be a result of the blockade of the PI3K/Akt/NF-κB signaling pathway.
Collapse
Affiliation(s)
- Wenjun Li
- Department of Pharmacy, The Third Affiliated Hospital of Chongqing Medical University (Gener Hospital), Chongqing, China
| | - Qian Du
- Department of Pharmacy, The Third Affiliated Hospital of Chongqing Medical University (Gener Hospital), Chongqing, China
| | - Xiaoli Li
- College of Pharmacy, Chongqing Medical University, Chongqing, China
| | - Xiangru Zheng
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Feng Lv
- Department of Pharmacy, The Third Affiliated Hospital of Chongqing Medical University (Gener Hospital), Chongqing, China
| | - Xin Xi
- Department of Pharmacy, The Third Affiliated Hospital of Chongqing Medical University (Gener Hospital), Chongqing, China
| | - Guili Huang
- Department of Pharmacy, The Third Affiliated Hospital of Chongqing Medical University (Gener Hospital), Chongqing, China
| | - Jia Yang
- Department of Pharmacy, The Third Affiliated Hospital of Chongqing Medical University (Gener Hospital), Chongqing, China
| | - Songqing Liu
- Department of Pharmacy, The Third Affiliated Hospital of Chongqing Medical University (Gener Hospital), Chongqing, China
| |
Collapse
|
76
|
From tea to treatment; epigallocatechin gallate and its potential involvement in minimizing the metabolic changes in cancer. Nutr Res 2019; 74:23-36. [PMID: 31918176 DOI: 10.1016/j.nutres.2019.12.004] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 12/02/2019] [Accepted: 12/06/2019] [Indexed: 01/09/2023]
Abstract
As the most abundant bioactive polyphenol in green tea, epigallocatechin gallate (EGCG) is a promising natural product that should be used in the discovery and development of potential drug leads. Due to its association with chemoprevention, EGCG may find a role in the development of therapeutics for prostate cancer. Natural products have long been used as a scaffold for drug design, as their already noted bioactivity can help accelerate the development of novel treatments. Green tea and the EGCG contained within have become associated with chemoprevention, and both in vitro and in vivo studies have correlated EGCG to inhibiting cell growth and increasing the metabolic stress of cancer cells, possibly giving merit to its long utilized therapeutic use in traditional therapies. There is accumulating evidence to suggest EGCG's role as an inhibitor of the phosphatidylinositol 3-kinase/protein kinase B/mammalian target of rapamycin signaling cascade, acting upon major axis points within cancer survival pathways. The purpose of this review is to examine the research conducted on tea along with EGCG in the areas of the treatment of and/or prevention of cancer. This review discusses Camellia sinensis as well as the bioactive phytochemical compounds contained within. Clinical uses of tea are explored, and possible pathways for activity are discussed before examining the evidence for EGCG's potential for acting on these processes. EGCG is identified as being a possible lead phytochemical for future drug design investigations.
Collapse
|
77
|
Wong KM, Song J, Saini V, Wong YH. Small Molecules as Drugs to Upregulate Metastasis Suppressors in Cancer Cells. Curr Med Chem 2019; 26:5876-5899. [PMID: 29788870 DOI: 10.2174/0929867325666180522090842] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Revised: 03/20/2018] [Accepted: 05/18/2018] [Indexed: 12/26/2022]
Abstract
It is well-recognized that the majority of cancer-related deaths is attributed to metastasis, which can arise from virtually any type of tumor. Metastasis is a complex multistep process wherein cancer cells must break away from the primary tumor, intravasate into the circulatory or lymphatic systems, extravasate, proliferate and eventually colonize secondary sites. Since these molecular processes involve the coordinated actions of numerous proteins, targeted disruptions of key players along these pathways represent possible therapeutic interventions to impede metastasis formation and reduce cancer mortality. A diverse group of proteins with demonstrated ability to inhibit metastatic colonization have been identified and they are collectively known as metastasis suppressors. Given that the metastasis suppressors are often downregulated in tumors, drug-induced re-expression or upregulation of these proteins represents a promising approach to limit metastasis. Indeed, over 40 compounds are known to exhibit efficacy in upregulating the expression of metastasis suppressors via transcriptional or post-transcriptional mechanisms, and the most promising ones are being evaluated for their translational potentials. These small molecules range from natural products to drugs in clinical use and they apparently target different molecular pathways, reflecting the diverse nature of the metastasis suppressors. In this review, we provide an overview of the different classes of compounds known to possess the ability to upregulate one or more metastasis suppressors, with an emphasis on their mechanisms of action and therapeutic potentials.
Collapse
Affiliation(s)
- Ka Ming Wong
- Division of Life Science and the Biotechnology Research Institute, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong
| | - Jiaxing Song
- Division of Life Science and the Biotechnology Research Institute, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong
| | - Vasu Saini
- Division of Life Science and the Biotechnology Research Institute, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong
| | - Yung H Wong
- Division of Life Science and the Biotechnology Research Institute, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong.,State Key Laboratory of Molecular Neuroscience, and the Molecular Neuroscience Center, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong.,Guangdong Provincial Key Laboratory of Brain Science, Disease and Drug Development, HKUST Shenzhen Research Institute, Shenzhen, China
| |
Collapse
|
78
|
Genistein potentiates Centchroman induced antineoplasticity in breast cancer via PI3K/Akt deactivation and ROS dependent induction of apoptosis. Life Sci 2019; 239:117073. [PMID: 31751581 DOI: 10.1016/j.lfs.2019.117073] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 10/28/2019] [Accepted: 11/14/2019] [Indexed: 12/30/2022]
Abstract
AIMS Recently, strategies of cancer treatment using combination of agents with distinct molecular mechanism(s) of action are considered more promising due to its high efficacy and reduced systemic toxicity. The study is aimed to improve the efficacy of selective estrogen receptor modulator, Centchroman (CC) by combination with the phytoestrogen Genistein (GN). METHODS Cytotoxicity was evaluated by Sulforhodamine B assay. Cell cycle analysis was done through flow cytometry. Further, Apoptosis was analyzed using Annexin V/PI staining, tunel assay and electron microscopic examination and verified using western blot analysis. In order to validate the in vitro results, in vivo analysis was performed using 4T1-syngeneic mouse model. KEY FINDINGS In this study, we report that the dietary isoflavone genistein (GN) synergistically improved antineoplasticity of CC in breast cancer by arresting cells at G2/M phase culminating in ROS dependent apoptosis. The combination of CC plus GN caused dysregulation of Bax and Bcl-2 ratio inducing mitochondrial dysfunction, activation of Caspase-3/7, -9 and PARP cleavage. Further, combination significantly suppresses phosphorylation of PI3K/Akt/NF-κB, enhancing apoptosis. Additionally, combination markedly reduced tumor growth compared to CC and GN alone in mouse 4T1 breast tumor model. SIGNIFICANCE Together, these studies suggest that GN represents a potential adjunct molecule whose role in CC induced apoptosis deserves attention.
Collapse
|
79
|
Li X, Wang F, Ren M, Du M, Zhou J. The effects of c-Src kinase on EMT signaling pathway in human lens epithelial cells associated with lens diseases. BMC Ophthalmol 2019; 19:219. [PMID: 31703690 PMCID: PMC6842207 DOI: 10.1186/s12886-019-1229-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Accepted: 10/28/2019] [Indexed: 12/18/2022] Open
Abstract
Background The signaling pathway of epithelial to mesenchymal transition (EMT) is regulated by c-Src kinase in many cells. The purpose of this study was to investigate the effects of c-Src kinase on EMT of human lens epithelial cells in vivo stimulated by different factors. Methods Human lens epithelial cells, HLE-B3, were exposed to either an inflammatory factor, specifically IL-1α, IL-6, TNF-α or IL-1β, at 10 ng/mL or high glucose (35.5 mM) for 30 mins. Activity of c-Src kinase was evaluated by the expression of p-Src418 with western blot assay. To investigate the effects of activation of c-Src on EMT, HLE-B3 cells were transfected with pCDNA3.1-SrcY530F to upregulate activity of c-Src kinase, and pSlience4.1-ShSrc to knock it down. The expressions of c-Src kinase and molecular markers of EMT such as E-cadherin, ZO-1, α-SMA, and Vimentin were examined at 48 h by RT-PCR and western blot. At 48 h and 72 h of transfection, cell proliferation was detected by MTT, and cell mobility and migration were determined by scratch and transwell assays. Results Activity of c-Src kinase, which causes the expression of p-Src418, was upregulated by different inflammatory factors and high glucose in HLE-B3 cells. When HLE-B3 cells were transfected with pCDNA3.1-SrcY530F, the expression of c-Src kinase was upregulated on both mRNA and protein levels, and activity of c-Src kinase, expression of p-Src418 increased. The expressions of both E-cadherin and ZO-1 were suppressed, while the expressions of vimentin and α-SMA were elevated on both mRNA and protein levels at the same time. Cell proliferation, mobility and migration increased along with activation of c-Src kinase. Conversely, when HLE-B3 cells were transfected with pSlience4.1-ShSrc, both c-Src kinase and p-Src418 expressions were knocked down. The expressions of E-cadherin and ZO-1 increased, but the expressions of Vimentin and α-SMA decreased; meanwhile, cell proliferation, mobility and migration reduced. Conclusions The c-Src kinase in lens epithelial cells is easily activated by external stimuli, resulting in the induction of cell proliferation, mobility, migration and EMT.
Collapse
Affiliation(s)
- Xingyu Li
- Department of Ophthalmology, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, Shaanxi, China
| | - Fang Wang
- Department of Ophthalmology, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, Shaanxi, China
| | - Meixia Ren
- Department of Ophthalmology, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, Shaanxi, China
| | - Minjuan Du
- Department of Ophthalmology, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, Shaanxi, China
| | - Jian Zhou
- Department of Ophthalmology, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, Shaanxi, China.
| |
Collapse
|
80
|
Dai J, Jiang C, Chen H, Chai Y. Rapamycin Attenuates High Glucose-Induced Inflammation Through Modulation of mTOR/NF-κB Pathways in Macrophages. Front Pharmacol 2019; 10:1292. [PMID: 31736762 PMCID: PMC6831745 DOI: 10.3389/fphar.2019.01292] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2019] [Accepted: 10/09/2019] [Indexed: 12/11/2022] Open
Abstract
Background: The NLRP3 inflammasome is one of the key contributors to impaired wound healing in diabetes. In this study, we assessed the role of rapamycin on high glucose-induced inflammation in THP-1-derived macrophages and investigated the underlying signaling mechanisms. Methods: THP-1-derived macrophages were treated with high glucose to induce NLRP3 inflammasome activation. The cells were pretreated with rapamycin, BAY 11-7082, or PDTC before exposure to HG. mTOR, NF-κB, and NLRP3 inflammasome expression were measured by western blotting. Results: We found that rapamycin reduced NLRP3 inflammasome activation in macrophages. Rapamycin reduced NLRP3 inflammasome activation by inhibiting mTOR phosphorylation and NF-κB activation. Moreover, mTOR siRNA inhibited NF-κB activation, leading to the suppression of NLRP3 inflammasome activation. Conclusion: Rapamycin can ameliorate high glucose-induced NLRP3 inflammasome activation by attenuating the mTOR/NF-κB signaling pathway in macrophages. Rapamycin may act as a possible therapeutic option for high glucose-induced inflammatory response in impaired wound healing in the future.
Collapse
Affiliation(s)
| | | | - Hua Chen
- Department of Orthopedic Surgery, Shanghai Jiao Tong University, Affiliated Sixth People’s Hospital, Shanghai, China
| | - Yimin Chai
- Department of Orthopedic Surgery, Shanghai Jiao Tong University, Affiliated Sixth People’s Hospital, Shanghai, China
| |
Collapse
|
81
|
Li H, Liu B, Wu J, Yu H, Huang H, Chen X, Chen B, Wu S, Ma J, Liu W, Chen X, Lan L, He Z, Zhang H. The inhibitory effect of tachyplesin I on thrombosis and its mechanisms. Chem Biol Drug Des 2019; 94:1672-1679. [PMID: 31108023 DOI: 10.1111/cbdd.13570] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 05/07/2019] [Accepted: 05/13/2019] [Indexed: 12/31/2022]
Abstract
Thrombotic diseases are major cause of cardiovascular diseases. This study was designed to investigate the effect of tachyplesin I on platelet aggregation and thrombosis. Platelet aggregation was analysed with a whole blood aggregometer. The mice were employed to investigate the effect of tachyplesin I on thrombosis in vivo. Tachyplesin I inhibited thrombin-induced platelet aggregation in a dose-dependent manner. Furthermore, tachyplesin I significantly reduced thrombosis in carrageenan-induced tail thrombosis model by intraperitoneal injection (0.1, 0.2 or 0.4 mg/kg) or intragastric administration (15, 30 or 60 mg/kg). Tachyplesin I also prolonged the bleeding time (BT) and clotting time (CT). The results revealed that tachyplesin I inhibited platelet aggregation and thrombosis by interfering the PI3K/AKT pathway. Tachyplesin I did not show significantly toxicity to mice under 300 mg/kg via intravenous injection. The results show that tachyplesin I inhibits thrombosis and has low toxicity. It is suggested that tachyplesin I has the potential to develop a new anti-thrombotic drug.
Collapse
Affiliation(s)
- Huimin Li
- Department of Biochemistry and Molecular Biology, Guangdong Medical University, Zhanjiang, China.,Guangdong Key Laboratory for Research and Development of Natural Drugs, Guangdong Medical University, Zhanjiang, China
| | - Bin Liu
- Department of Biochemistry and Molecular Biology, Guangdong Medical University, Zhanjiang, China
| | - Jun Wu
- Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Huajun Yu
- Department of Biochemistry and Molecular Biology, Guangdong Medical University, Zhanjiang, China.,Guangdong Key Laboratory for Research and Development of Natural Drugs, Guangdong Medical University, Zhanjiang, China
| | - Hui Huang
- Department of Biochemistry and Molecular Biology, Guangdong Medical University, Zhanjiang, China
| | - Xi Chen
- Department of Biochemistry and Molecular Biology, Guangdong Medical University, Zhanjiang, China
| | - Baoan Chen
- Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Shang Wu
- Department of Biochemistry and Molecular Biology, Guangdong Medical University, Zhanjiang, China
| | - Jingyao Ma
- Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Wen Liu
- Department of Biochemistry and Molecular Biology, Guangdong Medical University, Zhanjiang, China
| | - Xiaoyi Chen
- Department of Biochemistry and Molecular Biology, Guangdong Medical University, Zhanjiang, China
| | - Liubo Lan
- Department of Biochemistry and Molecular Biology, Guangdong Medical University, Zhanjiang, China
| | - Zhan He
- Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Haitao Zhang
- Department of Biochemistry and Molecular Biology, Guangdong Medical University, Zhanjiang, China.,Guangdong Key Laboratory for Research and Development of Natural Drugs, Guangdong Medical University, Zhanjiang, China
| |
Collapse
|
82
|
Qiu K, Xie Q, Jiang S, Lin T. Silencing of DJ-1 reduces proliferation, invasion, and migration of papillary thyroid cancer cells in vitro, probably by increase of PTEN expression. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2019; 12:2046-2055. [PMID: 31934026 PMCID: PMC6949646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 03/28/2019] [Indexed: 06/10/2023]
Abstract
AIMS To explore the function of DJ-1 on cell proliferation, migration, and invasion in human papillary thyroid carcinoma (PTC) cells. MATERIALS AND METHODS DJ-1 was knocked out by siRNA in K1 and TPC-1 cells and the efficiency of siRNA was examined by qRT-PCR and western blot. Cell proliferation, cell cycle, migration, and invasion were measured by CCK-8 assay, flow cytometry, colony formation assay and trans-well assay, respectively. RESULTS K1 and TPC-1 cells that were transfected with siRNA of DJ-1 had significantly lower expression levels of DJ-1 mRNA and protein. Down-regulation of DJ-1 significantly suppressed the cell proliferation, migration, and invasion. siRNA-mediated knock-down of DJ-1 increased the number of cells in the G0/G1 phase but reduced it in the S phase, while the G2/M phase was not affected. Moreover, the expression level of PTEN (Phosphatase and Tensin Homolog, PTEN) was found up-regulated in DJ-1-null cells. CONCLUSIONS This work suggested that DJ-1 implicated in cell proliferation, migration, and invasion of papillary thyroid cancer cells, possibly by the DJ-1/PTEN/PI3K/Akt signal pathway.
Collapse
Affiliation(s)
- Kai Qiu
- Department of Vascular and Thyroid Surgery, Fujian Medical University Union Hospital Fuzhou, Fujian, P. R. China
| | - Qingji Xie
- Department of Vascular and Thyroid Surgery, Fujian Medical University Union Hospital Fuzhou, Fujian, P. R. China
| | - Shan Jiang
- Department of Vascular and Thyroid Surgery, Fujian Medical University Union Hospital Fuzhou, Fujian, P. R. China
| | - Ting Lin
- Department of Vascular and Thyroid Surgery, Fujian Medical University Union Hospital Fuzhou, Fujian, P. R. China
| |
Collapse
|
83
|
Lian Z, Han J, Huang L, Wei C, Fan Y, Xu J, Zhou M, Feng H, Liu Q, Chen L, Li Z, Cheng H, Yuan G, Lin X, Song F, Su Y, Wang C, Huang G, Qin A, Song Y, Yao G. Retraction of: A005, a novel inhibitor of phosphatidylinositol 3-kinase/mammalian target of rapamycin, prevents osteosarcoma-induced osteolysis. Carcinogenesis 2019; 40:e1-e13. [PMID: 29635391 DOI: 10.1093/carcin/bgy036] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Osteosarcoma is the most frequent primary bone tumor in children and adolescents. The phosphatidylinositol 3-kinase (PI3K)/mammalian target of rapamycin (mTOR) signaling pathway is an attractive anticancer target because it plays key roles in the regulation of cell growth, division and differentiation. In this study, we demonstrated high expression of PI3K/mTOR signaling pathway-related genes in patients with osteosarcoma. We thus investigated the effects of A005, a newly synthesized dual PI3K/mTOR inhibitor, on osteosarcoma cells and in a mouse xenograft tumor model. The results confirmed that A005 inhibited the proliferation, migration and invasion of human osteosarcoma cells. In addition, A005 also inhibited receptor activator of nuclear factor kappa-B ligand (RANKL)-induced osteoclast differentiation and bone resorption in vitro. Therefore, A005 was further applied to a SaOS-2 osteosarcoma-induced mouse osteolysis model. A005 inhibited tumor growth and prevented osteosarcoma-associated osteolysis via modulation of the PI3K/AKT/mTOR pathway. Overall, our results showed that A005 inhibited osteoclastogenesis and prevented osteosarcoma-induced bone osteolysis by suppressing PI3K/AKT/mTOR signaling. These findings indicated that A005 may be a promising candidate drug for the treatment of human osteosarcoma.
Collapse
Affiliation(s)
- Zhen Lian
- Department of Orthopedics, The Second Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong, China
| | - Jinsong Han
- Department of Medicinal Chemistry, School of Pharmacy, Second Military Medical University, Shanghai, China
- Department of Medicinal Chemistry, Organisch-Chemisches Institut, Ruprecht-Karls-Universität Heidelberg, Im Neuenheimer Feld, Heidelberg, Germany
| | - Lin Huang
- Department of Spine Surgery, Department of Orthopedics, Research Center of Spinal and Pelvic Tumor, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou City, Guangdong Province, China
| | - Chengming Wei
- Department of Orthopedics, Collaborative Innovation Center of Guangxi Biological Medicine, Guangxi Medical University, Guangxi, China
| | - Yongyong Fan
- Orthopedic Department, Taizhou Hospital, Wenzhou Medical University, Linhai, China
| | - Jiake Xu
- Department of Orthopedics, School of Pathology and Laboratory Medicine, The University of Western Australia, Perth, Western Australia, Australia
| | - Mengyu Zhou
- Department of Stomatology, Haicheng Central Hospital, Liaoning, China
| | - Haotian Feng
- Department of Orthopedics, Research Centre for Regenerative Medicine, Guangxi Medical University, Guangxi, China
| | - Qian Liu
- Department of Orthopedics, Research Centre for Regenerative Medicine, Guangxi Medical University, Guangxi, China
| | - Lingzi Chen
- Department of Orthopedics, The Second Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong, China
| | - Zhaoning Li
- Department of Orthopedics, The Second Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong, China
| | - Haichun Cheng
- Department of General Surgery, The Shenzhen Shajing Affiliated Hospital of Guangzhou Medical University, Shenzhen, Guangdong, China
| | - Guixin Yuan
- Department of Orthopedics, The Second Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong, China
| | - Xixi Lin
- Department of Orthopedics, Research Centre for Regenerative Medicine, Guangxi Medical University, Guangxi, China
| | - Fangming Song
- Department of Orthopedics, Research Centre for Regenerative Medicine, Guangxi Medical University, Guangxi, China
| | - Yiji Su
- Department of Rehabilitation, The First Affiliated Hospital, Guangxi Medical University, Guangxi, China
| | - Chao Wang
- Department of Orthopedics, School of Pathology and Laboratory Medicine, The University of Western Australia, Perth, Western Australia, Australia
| | - Guopeng Huang
- Department of Orthopedics, The Second Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong, China
| | - An Qin
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopedics, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yunlong Song
- Department of Medicinal Chemistry, School of Pharmacy, Second Military Medical University, Shanghai, China
| | - Guanfeng Yao
- Department of Orthopedics, The Second Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong, China
| |
Collapse
|
84
|
Karimabad MN, Mahmoodi M, Jafarzadeh A, Darekordi A, Hajizadeh MR, Hassanshahi G. Molecular Targets, Anti-cancer Properties and Potency of Synthetic Indole-3-carbinol Derivatives. Mini Rev Med Chem 2019; 19:540-554. [DOI: 10.2174/1389557518666181116120145] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Revised: 04/25/2017] [Accepted: 04/26/2017] [Indexed: 12/14/2022]
Abstract
The indole-3-carbinol (I3C) displays anti-cancer/proliferative activities against human cancer cells. Cellular proliferation is an event associated with the progress and its continuation. This manifest is described by variation in expression and/or functions of genes that are related with cell cycle relevant proteins. The constitutive activation of several signal transduction pathways stimulates cells proliferation as well. The immediate stages in cancer development are accompanied by a fibrogenic response and the progression of the hypoxic environment is in favor of survival and proliferatory functions of cancer stem cells. A main part for prevention of in cancer cells death may manifest through altering cell metabolism. Cellular proliferation and metastasis are reported to be supported with increased generation of responsible hormones (in hormone dependent malignancies), and further promotion the angiogenesis, with epithelial to mesenchymal transition. This may be facilitated by progression of autophagy phenomenon, as well as via taking cues from neighboring stromal cells. Several signaling pathways in association with various factors specific for cellular viability, including hypoxia inducible factor 1, NF-κB, insulin-like growth factor 1 (IGF-1) receptor, Human foreskin fibroblasts (HFF-1), phosphoinositide 3 kinase/Akt, Wnt, cell cycle related protein, with androgen and estrogen receptor signaling are reported to be inhibited by I3C. These evidences, in association with bioinformatics data represent very important information for describing signaling pathways in parallel with molecular targets that may serve as markers for early diagnosis and/or critical targets for designing and development of novel therapeutic regimes alone or combined with drugs, to prevent tumor formation and further progression. In particular, I3C and DIM have been extensively investigated for their importance against numbers human cancers both in vitro and in vivo. We aimed the present manuscript, current study, to review anticancer properties and the miscellaneous mechanisms underlying the antitumorigenicity in an in-depth study for broadening the I3C treating marvel.
Collapse
Affiliation(s)
- Mojgan Noroozi Karimabad
- Molecular Medicine Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Mehdi Mahmoodi
- Molecular Medicine Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Abdolah Jafarzadeh
- Molecular Medicine Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Ali Darekordi
- Department of Chemistry, Faculty of Science, Vali-e-Asr University of Rafsanjan, Rafsanjan, Iran
| | - Mohamad Reza Hajizadeh
- Molecular Medicine Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Gholamhossein Hassanshahi
- Molecular Medicine Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| |
Collapse
|
85
|
Mortezaee K, Najafi M, Farhood B, Ahmadi A, Shabeeb D, Musa AE. NF‐κB targeting for overcoming tumor resistance and normal tissues toxicity. J Cell Physiol 2019; 234:17187-17204. [DOI: 10.1002/jcp.28504] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 02/22/2019] [Accepted: 03/05/2019] [Indexed: 12/13/2022]
Affiliation(s)
- Keywan Mortezaee
- Department of Anatomy School of Medicine, Kurdistan University of Medical Sciences Sanandaj Iran
| | - Masoud Najafi
- Radiology and Nuclear Medicine Department School of Paramedical Sciences, Kermanshah University of Medical Sciences Kermanshah Iran
| | - Bagher Farhood
- Departments of Medical Physics and Radiology Faculty of Paramedical Sciences, Kashan University of Medical Sciences Kashan Iran
| | - Amirhossein Ahmadi
- Pharmaceutical Sciences Research Center Faculty of Pharmacy, Mazandaran University of Medical Sciences Sari Iran
| | - Dheyauldeen Shabeeb
- Department of Physiology College of Medicine, University of Misan Misan Iraq
| | - Ahmed E. Musa
- Department of Medical Physics Tehran University of Medical Sciences (International Campus) Tehran Iran
| |
Collapse
|
86
|
Sun Y, Zhou QM, Lu YY, Zhang H, Chen QL, Zhao M, Su SB. Resveratrol Inhibits the Migration and Metastasis of MDA-MB-231 Human Breast Cancer by Reversing TGF-β1-Induced Epithelial-Mesenchymal Transition. Molecules 2019; 24:molecules24061131. [PMID: 30901941 PMCID: PMC6471699 DOI: 10.3390/molecules24061131] [Citation(s) in RCA: 105] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 03/17/2019] [Accepted: 03/19/2019] [Indexed: 01/20/2023] Open
Abstract
Metastasis is a major cause of death in patients with breast cancer. In the process of cancer development, epithelial-mesenchymal transition (EMT) is crucial to promoting the invasion and migration of tumor cells. In a previous study, the role of resveratrol in migration and metastasis was investigated in MDA-MB-231 (MDA231) human breast cancer cells and a xenograft-bearing mouse model. Additionally, the related mechanism was explored. In the present study, in vitro Transwell assays showed that resveratrol can inhibit the migration of transforming growth factor (TGF)-β1-induced MDA231 cells in a concentration-dependent manner. An enzyme-linked immunosorbent assay (ELISA) showed that resveratrol can reduce the secretion of matrix metalloproteinase (MMP)-2 and MMP-9. Immunofluorescence was performed to confirm the expression of EMT-related markers. Immunofluorescence assays confirmed that resveratrol changed the expression of the EMT-related markers E-cadherin and vimentin. Western blot analysis demonstrated that resveratrol decreased the expression levels of MMP-2, MMP-9, Fibronectin, α-SMA, P-PI3K, P-AKT, Smad2, Smad3, P-Smad2, P-Smad3, vimentin, Snail1, and Slug, as well as increased the expression levels of E-cadherin in MDA231 cells. In vivo, resveratrol inhibited lung metastasis in a mouse model bearing MDA231 human breast cancer xenografts without marked changes in body weight or liver and kidney function. These results indicate that resveratrol inhibits the migration of MDA231 cells by reversing TGF-β1-induced EMT and inhibits the lung metastasis of MDA231 human breast cancer in a xenograft-bearing mouse model.
Collapse
Affiliation(s)
- Yang Sun
- Research Center for Traditional Chinese Medicine Complexity System, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Qian-Mei Zhou
- Research Center for Traditional Chinese Medicine Complexity System, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Yi-Yu Lu
- Research Center for Traditional Chinese Medicine Complexity System, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Hui Zhang
- Research Center for Traditional Chinese Medicine Complexity System, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Qi-Long Chen
- Research Center for Traditional Chinese Medicine Complexity System, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Ming Zhao
- Research Center for Traditional Chinese Medicine Complexity System, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
- AntiCancer Inc., San Diego, CA 92100, USA.
| | - Shi-Bing Su
- Research Center for Traditional Chinese Medicine Complexity System, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| |
Collapse
|
87
|
Tang H, Bai Y, Pan G, Wang X, Wei Y, Yang Z, Zhao J. Interleukin-6 and insulin-like growth factor-1 synergistically promote the progression of NSCLC. Autoimmunity 2019; 51:399-407. [PMID: 30604632 DOI: 10.1080/08916934.2018.1550079] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Affiliation(s)
- Hexiao Tang
- Department of Thoracic and Cardiovascular Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yuquan Bai
- Department of Thoracic and Cardiovascular Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Gaofeng Pan
- Department of Thoracic and Cardiovascular Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Xianguo Wang
- Department of Thoracic and Cardiovascular Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yanhong Wei
- Department of Nephrology, Wuhan Central Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zetian Yang
- Department of Thoracic and Cardiovascular Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Jinping Zhao
- Department of Thoracic and Cardiovascular Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
88
|
Hseu YC, Lin YC, Rajendran P, Thigarajan V, Mathew DC, Lin KY, Way TD, Liao JW, Yang HL. Antrodia salmonea suppresses invasion and metastasis in triple-negative breast cancer cells by reversing EMT through the NF-κB and Wnt/β-catenin signaling pathway. Food Chem Toxicol 2018; 124:219-230. [PMID: 30529123 DOI: 10.1016/j.fct.2018.12.009] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Revised: 11/29/2018] [Accepted: 12/06/2018] [Indexed: 01/21/2023]
Abstract
Antrodia salonea (AS), a fungus that is indigenous to Taiwan has been well known for its anti-cancer properties. We investigated the anti-metastatic and anti-epithelial-mesenchymal transition (EMT) properties of AS in TNBC cells. To determine their EMT and metastasis levels, in vitro wound healing, wound invasion, Western blotting, RT-PCR, luciferase activity and immunofluorescence assays were performed, while the in vivo anti-metastatic efficacy of AS was evaluated in BALB/c-nu mice through bioluminescence imaging, HE staining, and immunohistochemical staining. MDA-MB-231 cells, when treated with AS concentrations (25-100 μg/mL) resulted in significant reduction of invasion and migration as well as the downregulation of VEGF, uPAR, uPA and MMP-9 (inhibition of PI3K/AKT/NFκB pathways). AS treatment prevented morphological changes and reversed EMT through the upregulation of E-cadherin and the downregulation of N-cadherin, Slug, Twist, and Vimentin. Inhibition of Smad3 signaling pathway, downregulation of β-catenin pathway and upregulation of GSK3β expression were also observed while, suppression of metastasis and EMT in TGF-β1-stimulated non-tumorigenic MCF-10A cells was observed when treated with AS. Histological analysis confirmed that AS reduced tumor metastasis and upregulated E-cadherin expression in biopsied lung tissues. Our results indicated that AS exhibits anti-EMT and anti-metastatic activity, that could contribute to develop anticancer drugs against TNBC.
Collapse
Affiliation(s)
- You-Cheng Hseu
- Department of Cosmeceutics, College of Biopharmaceutical and Food Sciences, China Medical University, Taichung, 40402, Taiwan; Department of Health and Nutrition Biotechnology, Asia University, Taichung, 41354, Taiwan; Chinese Medicine Research Center, China Medical University, Taichung, 40402, Taiwan
| | - Yi-Chun Lin
- Institute of Nutrition, College of Biopharmaceutical and Food Sciences, China Medical University, Taichung, 40402, Taiwan
| | - Peramaiyan Rajendran
- Department of Cosmeceutics, College of Biopharmaceutical and Food Sciences, China Medical University, Taichung, 40402, Taiwan
| | - Varadharajan Thigarajan
- Institute of Nutrition, College of Biopharmaceutical and Food Sciences, China Medical University, Taichung, 40402, Taiwan
| | - Dony Chacko Mathew
- Department of Cosmeceutics, College of Biopharmaceutical and Food Sciences, China Medical University, Taichung, 40402, Taiwan
| | - Kai-Yuan Lin
- Department of Medical Research, Chi-Mei Medical Center, Tainan, 710, Taiwan
| | - Tzong-Der Way
- Department of Life Sciences, College of Biopharmaceutical and Food Sciences, China Medical University, Taichung, 40402, Taiwan
| | - Jiunn-Wang Liao
- Graduate Institute of Veterinary Pathology, National Chung Hsing University, Taichung, 402, Taiwan
| | - Hsin-Ling Yang
- Institute of Nutrition, College of Biopharmaceutical and Food Sciences, China Medical University, Taichung, 40402, Taiwan.
| |
Collapse
|
89
|
Afshari K, Dehdashtian A, Haddadi NS, Haj-Mirzaian A, Iranmehr A, Ebrahimi MA, Tavangar SM, Faghir-Ghanesefat H, Mohammadi F, Rahimi N, Javidan AN, Dehpour AR. Anti-inflammatory effects of Metformin improve the neuropathic pain and locomotor activity in spinal cord injured rats: introduction of an alternative therapy. Spinal Cord 2018; 56:1032-1041. [PMID: 29959433 DOI: 10.1038/s41393-018-0168-x] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Revised: 05/28/2018] [Accepted: 05/29/2018] [Indexed: 12/15/2022]
Abstract
STUDY DESIGN This is an animal study. OBJECTIVES Metformin is a safe drug for controlling blood sugar in diabetes. It has been shown that metformin improves locomotor recovery after spinal cord injury (SCI). Neuropathic pain is also a disturbing component of SCI. It is indicated that metformin has neuroprotective and anti-inflammatory effects, which attenuate neuropathic pain and hyperalgesia in injured nerves. Thus, we evaluated metformin's therapeutic effects on SCI neuroinflammation and its sensory and locomotor complications. Meanwhile, results were compared to minocycline, an anti-neuroinflammation therapy in SCI. SETTING Experimental Medicine Research Center, Tehran University of Medical Sciences, Iran METHODS: In an animal model of SCI, 48 male rats were subjected to T9 vertebra laminectomy. Animals were divided into a SHAM-operated group and five treatment groups. The treatments included normal saline as a vehicle control group, minocycline 90 mg/kg and metformin at the doses of 10, 50 and 100 mg/kg. Locomotor scaling, behavioral tests for neuropathic pain and weight changes were evaluated and compared through a 28-days period. At the end of the study, tissue samples were taken to assess neuroinflammatory changes. RESULTS Metformin 50 mg/kg improved the locomotors ability (p < 0.001) and decreased sensitivity to mechanical and thermal allodynia (p < 0.01). These results were compatible with minocycline effect on SCI (p > 0.05). While metformin led to weight loss, both metformin and minocycline significantly decreased neuroinflammation in the assessment of cord tissue histopathology, and levels of TNF-α and interleukin-1β (p < 0.001). CONCLUSIONS Metformin could be considered as an alternative therapeutic agent for SCI, as it potentially attenuates neuroinflammation, sensory and locomotor complications of cord injury.
Collapse
Affiliation(s)
- Khashayar Afshari
- MD-MPH, Tehran University of Medical Sciences, Tehran, Iran
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran
- Brain and Spinal Cord Injury Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Amir Dehdashtian
- MD-MPH, Tehran University of Medical Sciences, Tehran, Iran
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Nazgol-Sadat Haddadi
- MD-MPH, Tehran University of Medical Sciences, Tehran, Iran
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Arvin Haj-Mirzaian
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran
- MD, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Arad Iranmehr
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran
- Neurosurgery Resident, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Ali Ebrahimi
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran
- Phar.D., Tehran University of Medical Sciences, Tehran, Iran
| | - Seyed Mohammad Tavangar
- Chronic Diseases Research Center, Endocrinology and Metabolism Population Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
- Professor of Pathology, Department of Pathology, Dr. Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Fatemeh Mohammadi
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran
- Brain and Spinal Cord Injury Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Nastaran Rahimi
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Abbas Norouzi Javidan
- Brain and Spinal Cord Injury Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
- Associate professor of Physiology, Ph.D. in Physiology, Tehran University of Medical Sciences, Tehran, Iran
| | - Ahmad Reza Dehpour
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran.
- Professor of Pharmacology, Ph.D. in Pharmacology, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
90
|
Yu W, Liang X, Li X, Zhang Y, Sun Z, Liu Y, Wang J. MicroRNA-195: a review of its role in cancers. Onco Targets Ther 2018; 11:7109-7123. [PMID: 30410367 PMCID: PMC6200091 DOI: 10.2147/ott.s183600] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
MicroRNAs (miRNAs) are small and highly conserved noncoding RNAs that regulate gene expression at the posttranscriptional level by binding to the 3′-UTR of target mRNAs. Recently, increasing evidence has highlighted their profound roles in various pathological processes, including human cancers. Deregulated miRNAs function as either oncogenes or tumor suppressor genes in multiple cancer types. Among them, miR-195 has been reported to significantly impact oncogenicity in various neoplasms by binding to critical genes and signaling pathways, enhancing or inhibiting the progression of cancers. In this review, we focus on the expression of miR-195 in regulatory mechanisms and tumor biological processes and discuss the future potential therapeutic implications of diverse types of human malignancies.
Collapse
Affiliation(s)
- Wanpeng Yu
- Institute for Translational Medicine, Medical College of Qingdao University, Qingdao 266021, China;
| | - Xiao Liang
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao 266109, China
| | - Xiangdong Li
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Yuan Zhang
- Institute for Translational Medicine, Medical College of Qingdao University, Qingdao 266021, China;
| | - Zhenqing Sun
- Department of General Surgery, The Affiliated Hospital of Qingdao University, Qingdao 266003, China
| | - Ying Liu
- Institute for Translational Medicine, Medical College of Qingdao University, Qingdao 266021, China;
| | - Jianxun Wang
- Institute for Translational Medicine, Medical College of Qingdao University, Qingdao 266021, China;
| |
Collapse
|
91
|
Nitulescu GM, Van De Venter M, Nitulescu G, Ungurianu A, Juzenas P, Peng Q, Olaru OT, Grădinaru D, Tsatsakis A, Tsoukalas D, Spandidos DA, Margina D. The Akt pathway in oncology therapy and beyond (Review). Int J Oncol 2018; 53:2319-2331. [PMID: 30334567 PMCID: PMC6203150 DOI: 10.3892/ijo.2018.4597] [Citation(s) in RCA: 163] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Accepted: 10/10/2018] [Indexed: 02/07/2023] Open
Abstract
Protein kinase B (Akt), similar to many other protein kinases, is at the crossroads of cell death and survival, playing a pivotal role in multiple interconnected cell signaling mechanisms implicated in cell metabolism, growth and division, apoptosis suppression and angiogenesis. Akt protein kinase displays important metabolic effects, among which are glucose uptake in muscle and fat cells or the suppression of neuronal cell death. Disruptions in the Akt-regulated pathways are associated with cancer, diabetes, cardiovascular and neurological diseases. The regulation of the Akt signaling pathway renders Akt a valuable therapeutic target. The discovery process of Akt inhibitors using various strategies has led to the identification of inhibitors with great selectivity, low side-effects and toxicity. The usefulness of Akt emerges beyond cancer therapy and extends to other major diseases, such as diabetes, heart diseases, or neurodegeneration. This review presents key features of Akt structure and functions, and presents the progress of Akt inhibitors in regards to drug development, and their preclinical and clinical activity in regards to therapeutic efficacy and safety for patients.
Collapse
Affiliation(s)
- George Mihai Nitulescu
- Faculty of Pharmacy, 'Carol Davila' University of Medicine and Pharmacy, 020956 Bucharest, Romania
| | - Maryna Van De Venter
- Department of Biochemistry and Microbiology, Nelson Mandela University, Port Elizabeth 6031, South Africa
| | - Georgiana Nitulescu
- Faculty of Pharmacy, 'Carol Davila' University of Medicine and Pharmacy, 020956 Bucharest, Romania
| | - Anca Ungurianu
- Faculty of Pharmacy, 'Carol Davila' University of Medicine and Pharmacy, 020956 Bucharest, Romania
| | - Petras Juzenas
- Department of Pathology, Radiumhospitalet, Oslo University Hospital, 0379 Oslo, Norway
| | - Qian Peng
- Department of Pathology, Radiumhospitalet, Oslo University Hospital, 0379 Oslo, Norway
| | - Octavian Tudorel Olaru
- Faculty of Pharmacy, 'Carol Davila' University of Medicine and Pharmacy, 020956 Bucharest, Romania
| | - Daniela Grădinaru
- Faculty of Pharmacy, 'Carol Davila' University of Medicine and Pharmacy, 020956 Bucharest, Romania
| | - Aristides Tsatsakis
- Department of Forensic Sciences and Toxicology, Faculty of Medicine, University of Crete, 71003 Heraklion, Greece
| | - Dimitris Tsoukalas
- Department of Forensic Sciences and Toxicology, Faculty of Medicine, University of Crete, 71003 Heraklion, Greece
| | - Demetrios A Spandidos
- Laboratory of Clinical Virology, School of Medicine, University of Crete, 71003 Heraklion, Greece
| | - Denisa Margina
- Faculty of Pharmacy, 'Carol Davila' University of Medicine and Pharmacy, 020956 Bucharest, Romania
| |
Collapse
|
92
|
Liu L, Wang H, Cui J, Zhang Q, Zhang W, Xu W, Lu H, Liu S, Shen S, Fang F, Li L, Yang W, Zhuang Z, Li J. Inhibition of Protein Phosphatase 2A Sensitizes Mucoepidermoid Carcinoma to Chemotherapy via the PI3K-AKT Pathway in Response to Insulin Stimulus. Cell Physiol Biochem 2018; 50:317-331. [PMID: 30282066 DOI: 10.1159/000494008] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Accepted: 09/25/2018] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND/AIMS Protein phosphatase 2A (PP2A) is a ubiquitous serine/threonine phosphatase that mediates cell cycle regulation and metabolism. Mounting evidence has indicated that PP2A inhibition exhibits considerable anticancer potency in multiple types of human cancers. However, the efficacy of PP2A inhibition remains unexplored in mucoepidermoid carcinoma (MEC), especially in locally advanced and metastatic cases with limited systemic treatment. In this study, we demonstrated the therapeutic potency of LB100 in mucoepidermoid carcinoma. METHODS In this study, the expression of PP2A was evaluated using immunohistochemical (IHC) staining. The effects associated with LB100 alone and in combination with cisplatin for the treatment of mucoepidermoid carcinoma were investigated both in vitro, regarding metabolism, proliferation, and migration, and in vivo in a mucoepidermoid carcinoma xenograft model. In addition, with LB100 treatment and in response to an insulin stimulus, the expression levels and phosphorylation levels of targets in the PI3K-AKT pathway were determined using western blot analysis and immunoblotting. RESULTS The expression of protein phosphatase 2A was significantly upregulated in the clinical specimens of high-grade MECs compared with those of low-/medium-grade MECs and normal controls. In this article, we report that a small molecule PP2A inhibitor, LB100, decreased cellular viability and glycolytic activity and induced G2/M cell cycle arrest. Importantly, LB100 enhanced the efficacy of cisplatin in mucoepidermoid carcinoma cells both in vitro and in vivo. PP2A inhibition by LB100 increased the phosphorylation of insulin receptor substrate 1(IRS-1) on serine residues, downregulated the expression of phosphatidylinositol 3-kinase (PI3K) p110 alpha subunit and dephosphorylated AKT at Ser473 and Thr308 in mucoepidermoid carcinoma cells in response to insulin stimulus. CONCLUSION These results highlight the translational potential of PP2A inhibition to synergize with cisplatin in mucoepidermoid carcinoma treatment.
Collapse
Affiliation(s)
- Limin Liu
- Department of Oral Pathology, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, National Clinical Research Center for Oral Diseases, Shanghai, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, China.,Neuro-Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Herui Wang
- Neuro-Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Jing Cui
- Neuro-Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Qi Zhang
- Neuro-Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Wei Zhang
- Neuro-Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Wanlin Xu
- Department of Oral Pathology, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, National Clinical Research Center for Oral Diseases, Shanghai, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, China
| | - Hao Lu
- Department of Oral Pathology, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, National Clinical Research Center for Oral Diseases, Shanghai, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, China
| | - Shengwen Liu
- Department of Oral Pathology, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, National Clinical Research Center for Oral Diseases, Shanghai, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, China
| | - Shukun Shen
- Department of Oral Pathology, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, National Clinical Research Center for Oral Diseases, Shanghai, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, China
| | | | - Lei Li
- Department of Oral Pathology, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, National Clinical Research Center for Oral Diseases, Shanghai, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, China
| | - Wenjun Yang
- Department of Oral Pathology, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, National Clinical Research Center for Oral Diseases, Shanghai, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, China
| | - Zhengping Zhuang
- Neuro-Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Jiang Li
- Department of Oral Pathology, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, National Clinical Research Center for Oral Diseases, Shanghai, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, China
| |
Collapse
|
93
|
Long MP, Wang HL, Luo YB, Yang JH. Targeting ROR1 inhibits epithelial to mesenchymal transition in human lung adenocarcinoma via mTOR signaling pathway. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2018; 11:4759-4770. [PMID: 31949551 PMCID: PMC6962910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Accepted: 09/19/2018] [Indexed: 06/10/2023]
Abstract
The receptor tyrosine kinase-like orphan receptor 1 (ROR1) is a type I surface transmembrane protein that contributes to progression of tumor-cell growth and metastasis. We and others have shown that the roles of ROR1 include inhibiting apoptosis, potentiating EGFR signaling, and inducing proliferation in lung cancer, but the roles and mechanisms of ROR1 in lung adenocarcinoma metastasis have not been elucidated. Here we chose four lung adenocarcinoma cell lines, PC9 (erlotinib-sensitive), PC9erlo (acquired erlotinib-resistant), NCI-H358 (partial erlotinib-resistant), and NCI-H1975 (erlotinib-resistant) as cell models to simulate the clinical situation. We found that ROR1 prompted epithelial to mesenchymal transition (EMT) by increasing the expression level of a key epithelial gene, E-cadherin, while decreasing the expression level of the key mesenchymal gene vimentin. Silencing ROR1 by siRNA significantly reduced the migration and invasion of lung adenocarcinoma cells in vitro and also significantly inhibited the phosphorylation of Akt (Ser473), mTOR (Ser2448), Raptor (Ser792) and p70S6K (Thr389) in all four cell lines. This strongly supports our proposal that ROR1 may play a central role in tumor progression and metastasis in lung adenocarcinoma through mTOR signaling, regardless of its EGFR-TKI sensitivity status.
Collapse
Affiliation(s)
- Ming-Peng Long
- School of Basic Medicine, Chengdu University of Traditional Chinese MedicineChengdu, Sichuan, PR China
| | - Hui-Li Wang
- School of Basic Medicine, Chengdu University of Traditional Chinese MedicineChengdu, Sichuan, PR China
| | - Yong-Bin Luo
- Department of Clinical Laboratory, The First People’s Hospital of KunmingKunming, Yunnan, PR China
| | - Jia-Hui Yang
- School of Basic Medicine, Chengdu University of Traditional Chinese MedicineChengdu, Sichuan, PR China
| |
Collapse
|
94
|
Zou M, Xu C, Li H, Zhang X, Fan W. 3,3'-Diindolylmethane suppresses ovarian cancer cell viability and metastasis and enhances chemotherapy sensitivity via STAT3 and Akt signaling in vitro and in vivo. Arch Biochem Biophys 2018:S0003-9861(18)30087-0. [PMID: 30040917 DOI: 10.1016/j.abb.2018.07.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Revised: 06/26/2018] [Accepted: 07/04/2018] [Indexed: 01/05/2023]
Abstract
Signal transducer and activator of transcription-3 (STAT3) protein is constitutively activated in ovarian cancer. The purpose of this study was to investigate the effects of 3,3'-diindolylmethane (DIM) on the regulation of STAT3 signaling and ovarian cancer cell viability, invasion, and sensitivity to chemotherapy. Ovarian cancer SKOV3 and A2780 cell lines were treated with various concentrations of DIM for different periods of time for assessment of cell viability as well as gene expression before and after knockdown of STAT3 expression using STAT3 shRNA. DIM treatment potently suppressed the viabilities of ovarian cancer cells. Consequently, DIM inhibited xenograft growth in nude mice. In addition, at the gene level, DIM inhibited phosphorylation of STAT3 and AKT proteins and expression of their downstream proteins. Moreover, knockdown of STAT3 expression significantly enhanced DIM antitumor activity and cisplatin sensitivity. Their combination suppressed the protein expression of survivin, Bcl-2, Mcl-1, HIF-1α, VEGF, and MMPs, but activated caspase-3. Taken together, the antitumor activity of DIM is via inhibition of the STAT3 and Akt signaling pathways. The combination of STAT3 knockdown with DIM treatment could be further evaluated as a therapeutic strategy for the treatment of advanced ovarian cancer.
Collapse
Affiliation(s)
- Minghua Zou
- Department of Oncology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, 400010, China.
| | - Changhua Xu
- Department of Oncology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, 400010, China.
| | - Hua Li
- Department of Oncology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, 400010, China.
| | - Xianquan Zhang
- Department of Oncology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, 400010, China.
| | - Weidong Fan
- Department of Oncology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, 400010, China.
| |
Collapse
|
95
|
Li J, Qi C, Liu X, Li C, Chen J, Shi M. Fibulin-3 knockdown inhibits cervical cancer cell growth and metastasis in vitro and in vivo. Sci Rep 2018; 8:10594. [PMID: 30006571 PMCID: PMC6045626 DOI: 10.1038/s41598-018-28906-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Accepted: 06/29/2018] [Indexed: 12/02/2022] Open
Abstract
To explore the function of fibulin-3 in cervical carcinoma malignant cell growth and metastasis, fibulin-3 expression in normal cervical tissue, cervical intraepithelial neoplasia (CIN), and cervical carcinoma were evaluated by immunohistochemistry. Quantitative real-time-polymerase chain reaction, western blotting, and immunocytochemistry were performed to assess the expression of fibulin-3 at mRNA and protein levels in different invasive clone sublines. Fibulin-3 shRNA and fibulin-3 cDNA were used to transfect the strongly and weakly invasive clone sublines. Using in vitro and in vivo functional assays, we investigated the effects of down-regulating and up-regulating fibulin-3 expression on the proliferation and invasion of different clone sublines. Epithelial mesenchymal transition (EMT) and its signaling pathways PI3K/AKT and ERK were studied carefully in lentiviral transfection systems. Fibulin-3 was upregulated in cervical carcinoma, and its overexpression was significantly related with malignant phenotype and poor prognosis of cervical carcinoma. Fibulin-3 promoted cervical cancer cell invasive capabilities by eliciting EMT and activating the PI3K-Akt-mTOR signal transduction pathway. Fibulin-3 could facilitate the process of cervical cancer development. The results presented here will help develop novel prognostic factors and possible therapeutic options for patients with cervical cancer.
Collapse
Affiliation(s)
- Juan Li
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital affiliated to Shandong University, Jinan, 250021, China
| | - Chen Qi
- Department of Obstetrics and Gynecology, Shan Xian Maternal and Child Care and family planning service center, Shan Xian, 274300, China
| | - Xia Liu
- Department of Obstetrics and Gynecology, Shan Xian Maternal and Child Care and family planning service center, Shan Xian, 274300, China
| | - Changzhong Li
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital affiliated to Shandong University, Jinan, 250021, China
| | - Jie Chen
- Department of Maternal and Child Health, School of Public Health, Shandong University, Jinan, 250012, China
| | - Min Shi
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital affiliated to Shandong University, Jinan, 250021, China.
| |
Collapse
|
96
|
Xie Y, Li B, Bu W, Gao L, Zhang Y, Lan X, Hou J, Xu Z, Chang S, Yu D, Xie B, Wang Y, Wang H, Zhang Y, Wu X, Zhu W, Shi J. Dihydrocelastrol exerts potent antitumor activity in mantle cell lymphoma cells via dual inhibition of mTORC1 and mTORC2. Int J Oncol 2018; 53:823-834. [PMID: 29901111 DOI: 10.3892/ijo.2018.4438] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Accepted: 05/16/2018] [Indexed: 11/06/2022] Open
Abstract
Mantle cell lymphoma (MCL) is a distinct and highly aggressive subtype of B-cell non-Hodgkin lymphoma. Dihydrocelastrol (DHCE) is a dihydro-analog of celastrol, which is isolated from the traditional Chinese medicinal plant Tripterygium wilfordii. The present study aimed to investigate the effects of DHCE treatment on MCL cells, and to determine the mechanism underlying its potent antitumor activity in vitro and in vivo using the Cell Counting kit-8 assay, clonogenic assay, apoptosis assay, cell cycle analysis, immunofluorescence staining, western blotting and tumor xenograft models. The results demonstrated that DHCE treatment exerted minimal cytotoxic effects on normal cells, but markedly suppressed MCL cell proliferation by inducing G0/G1 phase cell cycle arrest, and inhibited MCL cell viability by stimulating apoptosis via extrinsic and intrinsic pathways. In addition, the results revealed that DHCE suppressed cell growth and proliferation by inhibiting mammalian target of rapamycin complex (mTORC)1-mediated phosphorylation of ribosomal protein S6 kinase and eukaryotic initiation factor 4E binding protein. Simultaneously, DHCE induced apoptosis and inhibited cell survival by suppressing mTORC2-mediated phosphorylation of protein kinase B and nuclear factor-κB activity. In addition to in vitro findings, DHCE treatment reduced the MCL tumor burden in a xenograft mouse model, without indications of toxicity. Furthermore, combined treatment with DHCE and bortezomib, a proteasome inhibitor, induced a synergistic cytotoxic effect on MCL cells. These findings indicated that DHCE may have the potential to serve as a novel therapeutic agent for the treatment of MCL through dually inhibiting mTORC1 and mTORC2.
Collapse
Affiliation(s)
- Yongsheng Xie
- Department of Hematology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, P.R. China
| | - Bo Li
- CAS Key Laboratory of Receptor Research, Drug Discovery and Design Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, P.R. China
| | - Wenxuan Bu
- Department of Hematology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, P.R. China
| | - Lu Gao
- Department of Hematology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, P.R. China
| | - Yong Zhang
- CAS Key Laboratory of Receptor Research, Drug Discovery and Design Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, P.R. China
| | - Xiucai Lan
- Department of Hematology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, P.R. China
| | - Jun Hou
- Department of Hematology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, P.R. China
| | - Zhijian Xu
- CAS Key Laboratory of Receptor Research, Drug Discovery and Design Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, P.R. China
| | - Shuaikang Chang
- Department of Hematology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, P.R. China
| | - Dandan Yu
- Department of Hematology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, P.R. China
| | - Bingqian Xie
- Department of Hematology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, P.R. China
| | - Yingcong Wang
- Department of Hematology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, P.R. China
| | - Houcai Wang
- Department of Hematology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, P.R. China
| | - Yiwen Zhang
- Department of Hematology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, P.R. China
| | - Xiaosong Wu
- Department of Hematology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, P.R. China
| | - Weiliang Zhu
- CAS Key Laboratory of Receptor Research, Drug Discovery and Design Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, P.R. China
| | - Jumei Shi
- Department of Hematology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, P.R. China
| |
Collapse
|
97
|
Lu J, Wang SL, Wang YC, Wu YN, Yu X, Zhao WZ, Wang JH. High WAVE3 expression correlates with proliferation, migration and invasion in human ovarian cancer. Oncotarget 2018; 8:41189-41201. [PMID: 28476025 PMCID: PMC5522302 DOI: 10.18632/oncotarget.17141] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2016] [Accepted: 03/01/2017] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND Wiskott-Aldrich syndrome verprolin-homologous (WAVE) 3, a member of the WASP/WAVE family of proteins, plays a critical role in cell motility and acts as an oncogene in some human cancers, but no sufficient information available to illustrate its involvement in ovarian cancer tumorigenesis and progression. METHODS The expression of WAVE3 in human ovarian cancer and normal tissue was analyzed by immunohistochemistry. WAVE3 gene and protein expression in different human ovarian cancer cell lines was tested by RT-PCR and western blotting. Stable cells of WAVE3-knockdown in SKOV3 cells or transfected high expression in A2780 cells were constructed. The WAVE3 expression and its correlation with MMPs, p38 MAPK and other factors were studied. The relationship between WAVE3 and oncogenicity in vivo was also evaluated by nude mice xenograft model. RESULTS Immunohistochemistry staining showed the highest WAVE3 expression in ovarian cancer metastases, high in ovarian cancer and weak in normal. In different cell lines, SKOV3 cells showed the highest WAVE3 expression, A2780 cells expressed the lowest. Elevated WAVE3 expression in A2780 cells promoted proliferation and decreased apoptosis, increased the cell number in G2/M phase and promoted migration significantly. Correspondingly, knockdown of WAVE3 in SKOV3 cells showed opposite effects. The WAVE3 expression showed positive correlation with MMPs, NF-κB, COX-2, VEGF and phospho-p38 MAPK, but not p38. The high expression of WAVE3 promoted tumorigenesis in vivo. CONCLUSIONS Our results suggested that WAVE3 may be pivotal in ovarian cancer cell motility, invasion and oncogenesis, which might be related with MMPs production and p38 MAPK pathway.
Collapse
Affiliation(s)
- Jin Lu
- Department of Gynecological Oncology Surgery, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing Medical University Affiliated Cancer Hospital, Nanjing 210036, China
| | - Su-Li Wang
- Department of Gynecological Oncology Surgery, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing Medical University Affiliated Cancer Hospital, Nanjing 210036, China
| | - Ying-Chun Wang
- Department of Gynecological Oncology Surgery, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing Medical University Affiliated Cancer Hospital, Nanjing 210036, China
| | - Yi-Nan Wu
- Department of Gynecological Oncology Surgery, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing Medical University Affiliated Cancer Hospital, Nanjing 210036, China
| | - Xi Yu
- The Nanjing Han & Zaenker Cancer Institute, OG Pharmaceuticals, Nanjing 210036, China
| | - Wan-Zhou Zhao
- The Nanjing Han & Zaenker Cancer Institute, OG Pharmaceuticals, Nanjing 210036, China
| | - Jin-Hua Wang
- Department of Gynecological Oncology Surgery, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing Medical University Affiliated Cancer Hospital, Nanjing 210036, China.,Jinling Hospital, Nanjing University, Nanjing 210036, China
| |
Collapse
|
98
|
Jin M, Li C, Zhang Q, Xing S, Kan X, Wang J. Effects of aspirin on proliferation, invasion and apoptosis of Hep-2 cells via the PTEN/AKT/NF-κB/survivin signaling pathway. Oncol Lett 2018; 15:8454-8460. [PMID: 29805582 PMCID: PMC5950550 DOI: 10.3892/ol.2018.8377] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Accepted: 01/03/2018] [Indexed: 12/18/2022] Open
Abstract
Aspirin may exhibit antitumor activities, as it is able to inhibit cell proliferation. However, the ability of aspirin to inhibit cellular proliferation in Hep-2 cells and its underlying molecular mechanisms have been poorly determined. The aim of the present study was to investigate whether aspirin may induce cell apoptosis in the neoplastic cell line Hep-2. The effects of aspirin on the migratory and invasive abilities of Hep-2 cells were also investigated using Transwell assays. In the present study, it was demonstrated that aspirin induced apoptosis and inhibited proliferation, migration and invasion in Hep-2 cells. Aspirin also significantly decreased the expression of B-cell lymphoma 2 (Bcl-2) and caspase-3, and increased the expression of Bcl-2-associated X protein, suggesting that aspirin induced apoptosis through the intrinsic apoptotic pathway. Hep-2 cells treated with aspirin exhibited a significant upregulation of phosphatase and tensin homolog (PTEN) and decreased levels of phosphorylated protein kinase B (AKT). However, the total amount of AKT protein was not altered in response to aspirin treatment. Furthermore, the expression of nuclear factor (NF)-κB and survivin, which are the downstream targets of the PTEN/AKT signaling pathway, was inhibited. These results indicated that the molecular mechanism underlying the antitumor effects of aspirin may be associated with the inhibition of tumor invasion and induction of apoptosis by regulating the activity of the PTEN/AKT/NF-κB/survivin signaling pathway.
Collapse
Affiliation(s)
- Mingji Jin
- Department of Pharmacy, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Chunyu Li
- Department of Otolaryngology, Daqing Longnan Hospital, Daqing, Heilongjiang 163453, P.R. China
| | - Qiang Zhang
- Department of Pharmacy, The Second Hospital of Heilongjiang, Harbin, Heilongjiang 150010, P.R. China
| | - Shu Xing
- Department of Pharmacy, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Xuan Kan
- Department of Otolaryngology, Head and Neck Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150010, P.R. China
| | - Jiayu Wang
- Department of Otolaryngology, The First Hospital of Harbin, Harbin, Heilongjiang 150010, P.R. China
| |
Collapse
|
99
|
Huang W, Wen J, Lin R, Wei P, Huang F. Effects of mTOR/NF‐κB signaling pathway and high thoracic epidural anesthesia on myocardial ischemia‐reperfusion injury via autophagy in rats. J Cell Physiol 2018; 233:6669-6678. [DOI: 10.1002/jcp.26320] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2017] [Accepted: 11/30/2017] [Indexed: 01/21/2023]
Affiliation(s)
- Wei‐Qiang Huang
- Cardio‐Cerebrovascular Disease Precision Medical Key Laboratory Cultivation Base of GuangxiDepartment of Geriatric CardiologyThe First Affiliated Hospital of Guangxi Medical UniversityNanningChina
| | - Jian‐Lin Wen
- Cardio‐Cerebrovascular Disease Precision Medical Key Laboratory Cultivation Base of GuangxiDepartment of Geriatric CardiologyThe First Affiliated Hospital of Guangxi Medical UniversityNanningChina
| | - Ri‐Qi Lin
- Cardio‐Cerebrovascular Disease Precision Medical Key Laboratory Cultivation Base of GuangxiDepartment of Geriatric CardiologyThe First Affiliated Hospital of Guangxi Medical UniversityNanningChina
| | - Peng Wei
- Cardio‐Cerebrovascular Disease Precision Medical Key Laboratory Cultivation Base of GuangxiDepartment of Geriatric CardiologyThe First Affiliated Hospital of Guangxi Medical UniversityNanningChina
| | - Feng Huang
- Cardio‐Cerebrovascular Disease Precision Medical Key Laboratory Cultivation Base of GuangxiDepartment of CardiologyThe First Affiliated Hospital of Guangxi Medical UniversityNanningChina
| |
Collapse
|
100
|
Ji C, Guo H, Zhang P, Kuang W, Fan Y, Wu L. AnnexinA5 promote glioma cell invasion and migration via the PI3K/Akt/NF-κB signaling pathway. J Neurooncol 2018. [PMID: 29520611 DOI: 10.1007/s11060-018-2818-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
As an important member of the Annexins, AnnexinA5 has been attributed important functions in trophoblast membrane repair, anticoagulation and cellular signal transduction. Accumulated studies show that AnnexinA5 is closely associated with various types of carcinomas. However, the potential contribution of AnnexinA5 to glioma cancer progression remains unclear. In this study, we report that AnnexinA5 is significantly upregulated in both high-grade glioma samples and glioma cell lines. Moreover, overexpression of AnnexinA5 promotes cell migration and invasion in vitro and tumorigenicity of glioma cells in nude mice, while knockdown of AnnexinA5 manifests a repressive function during these cellular processes. Importantly, mechanistic studies further reveal that AnnexinA5 is an essential transcriptional target of Snail via activating the PI3K/Akt/NF-κB signaling pathway. Taken together, these findings suggest that AnnexinA5 or the PI3K/Akt/NF-κB pathway may be promising therapeutic molecules to eradicate glioma metastases.
Collapse
Affiliation(s)
- Chenxing Ji
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Hua Guo
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Pei Zhang
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Wei Kuang
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Yanghua Fan
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Lei Wu
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China.
| |
Collapse
|