1
|
Cooney LN, O’Shea KD, Winfield HJ, Cahill MM, Pierce LT, McCarthy FO. Bisindolyl Maleimides and Indolylmaleimide Derivatives-A Review of Their Synthesis and Bioactivity. Pharmaceuticals (Basel) 2023; 16:1191. [PMID: 37764999 PMCID: PMC10534823 DOI: 10.3390/ph16091191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 08/01/2023] [Accepted: 08/02/2023] [Indexed: 09/29/2023] Open
Abstract
The evolution of bisindolyl maleimides and indolyl maleimide derivatives and their unique biological activities have stimulated great interest in medicinal chemistry programs. Bisindolylmaleimide (BIM)-type compounds arise from natural sources such as arcyriarubin and are biosynthetically related to indolocarbazoles. BIMs are commonly the immediate synthetic precursors of indolocarbazoles, lacking a central bond between the two aromatic units and making them more flexible and drug-like. Synthetic endeavours within this class of compounds are broad and have led to the development of both remarkably potent and selective protein kinase inhibitors. Clinical BIM examples include ruboxistaurin and enzastaurin, which are highly active inhibitors of protein kinase C-β. While BIMs are widely recognised as protein kinase inhibitors, other modes of activity have been reported, including the inhibition of calcium signalling and antimicrobial activity. Critically, structural differences can be used to exploit new bioactivity and therefore it is imperative to discover new chemical entities to address new targets. BIMs can be highly functionalised or chemically manipulated, which provides the opportunity to generate new derivatives with unique biological profiles. This review will collate new synthetic approaches to BIM-type compounds and their associated bioactivities with a focus on clinical applications.
Collapse
Affiliation(s)
| | | | | | | | | | - Florence O. McCarthy
- School of Chemistry and ABCRF, University College Cork, Western Road, T12K8AF Cork, Ireland; (L.N.C.); (K.D.O.); (H.J.W.); (M.M.C.); (L.T.P.)
| |
Collapse
|
2
|
Ang D, Kendall R, Atamian HS. Virtual and In Vitro Screening of Natural Products Identifies Indole and Benzene Derivatives as Inhibitors of SARS-CoV-2 Main Protease (M pro). BIOLOGY 2023; 12:biology12040519. [PMID: 37106720 PMCID: PMC10135783 DOI: 10.3390/biology12040519] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 03/25/2023] [Accepted: 03/26/2023] [Indexed: 04/29/2023]
Abstract
The rapid spread of the coronavirus disease 2019 (COVID-19) resulted in serious health, social, and economic consequences. While the development of effective vaccines substantially reduced the severity of symptoms and the associated deaths, we still urgently need effective drugs to further reduce the number of casualties associated with SARS-CoV-2 infections. Machine learning methods both improved and sped up all the different stages of the drug discovery processes by performing complex analyses with enormous datasets. Natural products (NPs) have been used for treating diseases and infections for thousands of years and represent a valuable resource for drug discovery when combined with the current computation advancements. Here, a dataset of 406,747 unique NPs was screened against the SARS-CoV-2 main protease (Mpro) crystal structure (6lu7) using a combination of ligand- and structural-based virtual screening. Based on 1) the predicted binding affinities of the NPs to the Mpro, 2) the types and number of interactions with the Mpro amino acids that are critical for its function, and 3) the desirable pharmacokinetic properties of the NPs, we identified the top 20 candidates that could potentially inhibit the Mpro protease function. A total of 7 of the 20 top candidates were subjected to in vitro protease inhibition assay and 4 of them (4/7; 57%), including two beta carbolines, one N-alkyl indole, and one Benzoic acid ester, had significant inhibitory activity against Mpro protease. These four NPs could be developed further for the treatment of COVID-19 symptoms.
Collapse
Affiliation(s)
- Dony Ang
- Computational and Data Sciences Program, Chapman University, Orange, CA 92866, USA
- Schmid College of Science and Technology, Chapman University, Orange, CA 92866, USA
| | - Riley Kendall
- Computational and Data Sciences Program, Chapman University, Orange, CA 92866, USA
- Schmid College of Science and Technology, Chapman University, Orange, CA 92866, USA
| | - Hagop S Atamian
- Schmid College of Science and Technology, Chapman University, Orange, CA 92866, USA
- Biological Sciences Program, Chapman University, Orange, CA 92866, USA
| |
Collapse
|
3
|
Li S, Yin S, Ding H, Shao Y, Zhou S, Pu W, Han L, Wang T, Yu H. Polyphenols as potential metabolism mechanisms regulators in liver protection and liver cancer prevention. Cell Prolif 2023; 56:e13346. [PMID: 36229407 DOI: 10.1111/cpr.13346] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 09/19/2022] [Accepted: 09/29/2022] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND Liver cancer is one of the common malignancies. The dysregulation of metabolism is a driver of accelerated tumourigenesis. Metabolic changes are well documented to maintain tumour growth, proliferation and survival. Recently, a variety of polyphenols have been shown to have a crucial role both in liver disease prevention and metabolism regulation. METHODS We conducted a literature search and combined recent data with systematic analysis to comprehensively describe the molecular mechanisms that link polyphenols to metabolic regulation and their contribution in liver protection and liver cancer prevention. RESULTS Targeting metabolic dysregulation in organisms prevents and resists the development of liver cancer, which has important implications for identifying new therapeutic strategies for the management and treatment of cancer. Polyphenols are a class of complex compounds composed of multiple phenolic hydroxyl groups and are the main active ingredients of many natural plants. They mediate a broad spectrum of biological and pharmacological functions containing complex lipid metabolism, glucose metabolism, iron metabolism, intestinal flora imbalance, as well as the direct interaction of their metabolites with key cell-signalling proteins. A large number of studies have found that polyphenols affect the metabolism of organisms by interfering with a variety of intracellular signals, thereby protecting the liver and reducing the risk of liver cancer. CONCLUSION This review systematically illustrates that various polyphenols, including resveratrol, chlorogenic acid, caffeic acid, dihydromyricetin, quercetin, catechins, curcumin, etc., improve metabolic disorders through direct or indirect pathways to protect the liver and fight liver cancer.
Collapse
Affiliation(s)
- Shuangfeng Li
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Haihe Laboratory of Modern Chinese Medicine, Tianjin, China
| | - Shuangshuang Yin
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Haihe Laboratory of Modern Chinese Medicine, Tianjin, China
| | - Hui Ding
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yingying Shao
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Haihe Laboratory of Modern Chinese Medicine, Tianjin, China
| | - Shiyue Zhou
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Haihe Laboratory of Modern Chinese Medicine, Tianjin, China
| | - Weiling Pu
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Haihe Laboratory of Modern Chinese Medicine, Tianjin, China
| | - Lifeng Han
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Haihe Laboratory of Modern Chinese Medicine, Tianjin, China
| | - Tao Wang
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Haihe Laboratory of Modern Chinese Medicine, Tianjin, China
| | - Haiyang Yu
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Haihe Laboratory of Modern Chinese Medicine, Tianjin, China
| |
Collapse
|
4
|
Wang L, Zhang Y, Xu Z, Li J, Zhu W. Total synthesis of the indolocarbazole alkaloid ZHD-0501 and its seven isomers. Org Chem Front 2022. [DOI: 10.1039/d2qo00844k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
An indolocarbazole alkaloid, ZHD-0501 (1), and its 7 stereoisomers (2–8) were totally synthesized from d/l-glucose and 2,3-dibromomaleimide in 22 step reactions, and the absolute configuration of ZHD-0501 was confirmed for the first time.
Collapse
Affiliation(s)
- Liping Wang
- Key Laboratory of Marine Drugs, Ministry of Education of China, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China
- Key Laboratory of Chemistry for Natural Products of Guizhou Province, Chinese Academy of Sciences, Guiyang 550014, China
| | - Yapeng Zhang
- Key Laboratory of Marine Drugs, Ministry of Education of China, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
| | - Zhihong Xu
- Key Laboratory of Marine Drugs, Ministry of Education of China, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
| | - Jing Li
- Key Laboratory of Marine Drugs, Ministry of Education of China, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
| | - Weiming Zhu
- Key Laboratory of Marine Drugs, Ministry of Education of China, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China
- Laboratory for Marine Drugs and Bioproducts, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao 266237, China
| |
Collapse
|
5
|
Luo Y, Yin S, Lu J, Zhou S, Shao Y, Bao X, Wang T, Qiu Y, Yu H. Tumor microenvironment: a prospective target of natural alkaloids for cancer treatment. Cancer Cell Int 2021; 21:386. [PMID: 34284780 PMCID: PMC8290600 DOI: 10.1186/s12935-021-02085-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Accepted: 07/08/2021] [Indexed: 12/17/2022] Open
Abstract
Malignant tumor has become one of the major diseases that seriously endangers human health. Numerous studies have demonstrated that tumor microenvironment (TME) is closely associated with patient prognosis. Tumor growth and progression are strongly dependent on its surrounding tumor microenvironment, because the optimal conditions originated from stromal elements are required for cancer cell proliferation, invasion, metastasis and drug resistance. The tumor microenvironment is an environment rich in immune/inflammatory cells and accompanied by a continuous, gradient of hypoxia and pH. Overcoming immunosuppressive environment and boosting anti-tumor immunity may be the key to the prevention and treatment of cancer. Most traditional Chinese medicine have been proved to have good anti-tumor activity, and they have the advantages of better therapeutic effect and few side effects in the treatment of malignant tumors. An increasing number of studies are giving evidence that alkaloids extracted from traditional Chinese medicine possess a significant anticancer efficiency via regulating a variety of tumor-related genes, pathways and other mechanisms. This paper reviews the anti-tumor effect of alkaloids targeting tumor microenvironment, and further reveals its anti-tumor mechanism through the effects of alkaloids on different components in tumor microenvironment.
Collapse
Affiliation(s)
- Yanming Luo
- Tianjin State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Shuangshuang Yin
- Tianjin State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Jia Lu
- Tianjin State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Shiyue Zhou
- Tianjin State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Yingying Shao
- Tianjin State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Xiaomei Bao
- Tianjin State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Tao Wang
- Tianjin State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Yuling Qiu
- School of Pharmacy, Tianjin Medical University, Tianjin, 300070, China.
| | - Haiyang Yu
- Tianjin State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China.
| |
Collapse
|
6
|
Wang Z, Hui C, Xie Y. Natural STAT3 inhibitors: A mini perspective. Bioorg Chem 2021; 115:105169. [PMID: 34333418 DOI: 10.1016/j.bioorg.2021.105169] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 07/03/2021] [Accepted: 07/09/2021] [Indexed: 12/22/2022]
Abstract
Signal transducer and activator of transcription 3 (STAT3) plays pivotal role in several cellular processes such as cell proliferation and survival and has been found to be aberrantly activated in many cancers. STAT3 is largely believed to be one of the key oncogenes and crucial therapeutic targets. Much research has suggested the leading mechanisms for regulating the STAT3 pathway and its role in promoting tumorigenesis. Therefore, intensive efforts have been devoted to develop potent STAT3 inhibitors and several of them are currently undergoing clinical trials. Nevertheless, many natural products were identified as STAT3 inhibitors but attract less attention compared to the small molecule counterpart. In this review, the development of natural STAT3 inhibitors with an emphasis on their biological profile and chemical synthesis are detailed. The current state of STAT3 inhibitors and the future directions and opportunities for STAT3 inhibitor are discussed.
Collapse
Affiliation(s)
- Zhuo Wang
- Southern University of Science and Technology, School of Medicine, Shenzhen 518055, People's Republic of China.
| | - Chunngai Hui
- Division of Life Science, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| | - Yusheng Xie
- Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan 250012, China
| |
Collapse
|
7
|
Chen L, Feng D, Qian Y, Cheng X, Song H, Qian Y, Zhang X, Wu Y, Lv H, Liu Q, Cheng G, Yang B, Gu M. Valtrate as a novel therapeutic agent exhibits potent anti-pancreatic cancer activity by inhibiting Stat3 signaling. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2021; 85:153537. [PMID: 33744595 DOI: 10.1016/j.phymed.2021.153537] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 02/09/2021] [Accepted: 03/01/2021] [Indexed: 06/12/2023]
Abstract
BACKGROUND Valtrate is a novel epoxy iridoid ester isolated from Chinese herbal medicine Valeriana jatamansi Jones with anti-proliferative activity against various human cancer cell lines. However, its efficacy and molecular mechanisms against pancreatic cancer (PC) cells are largely unclear. PURPOSE To investigate the anti-cancer effects of valtrate on PC cell lines and its underlying mechanisms. METHODS MTT assay was first performed to detect the effect of valtrate on cell viability in human PC cell lines and normal pancreatic epithelial cells HPDE. Cell apoptosis and cycle phase assay were detected by flow cytometry. The relative mRNA expressions of Bax, Bcl-2, c-Myc, and CyclinB1 were tested by quantitative PCR (qPCR) assay. The expression of relative proteins was detected by Western blotting (WB). A PANC-1luc cells xenograft mouse model in nu/nu female mice was used to elucidate the effect of valtrate on tumor growth in vivo. RESULTS Valtrate significantly inhibited the growth of PC cells without affecting the growth of normal pancreatic epithelial cells HPDE, induced significant apoptosis and cell cycle arrest in G2/M phase. Moreover, valtrate inhibited the tumor growth of PC cell PANC-1 in xenograft mice by 61%. Further mechanism study demonstrated that valtrate could increase the expression level of Bax, suppress Bcl-2 as well as c-Myc and Cyclin B1, inhibit the transcriptional activity of Stat3, while valtrate decreased the expression level of Stat3 and phosphated-Stat3 (Tyr705) and induced the high molecular aggregation of Stat3. Molecular docking analysis predicted that valtrate might interact with Cys712 of Stat3 protein. Valtrate could also induce a transient depleted intracellular glutathione (GSH) level and increased reactive oxygen species (ROS). NAC (N-acetylcysteine), a reducer reversed valtrate-induced the depletion of Stat3, p-Stat3, c-Myc, and Cyclin B1. CONCLUSION Valtrate exerts anti-cancer activity against PC cells by directly targeting Stat3 through a covalent linkage to inhibit Stat3 activity, which causes apoptosis and cell cycle arrest.
Collapse
Affiliation(s)
- Liping Chen
- College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China; Academy of Chinese Medical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
| | - Di Feng
- College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
| | - Yafang Qian
- The First Affiliation Hospital, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310006, China
| | - Xiao Cheng
- Huzhou Institute for Food and Drug Control, Huzhou 313000, China
| | - Huizhu Song
- College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
| | - Yifan Qian
- College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
| | - Xu Zhang
- College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
| | - Yali Wu
- College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
| | - Huawei Lv
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Qi Liu
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Gang Cheng
- College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China.
| | - Bo Yang
- College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China.
| | - Mancang Gu
- College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China; Academy of Chinese Medical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China.
| |
Collapse
|
8
|
Garg M, Shanmugam MK, Bhardwaj V, Goel A, Gupta R, Sharma A, Baligar P, Kumar AP, Goh BC, Wang L, Sethi G. The pleiotropic role of transcription factor STAT3 in oncogenesis and its targeting through natural products for cancer prevention and therapy. Med Res Rev 2020; 41:1291-1336. [PMID: 33289118 DOI: 10.1002/med.21761] [Citation(s) in RCA: 80] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 10/30/2020] [Accepted: 11/18/2020] [Indexed: 12/11/2022]
Abstract
Signal transducer and activator of transcription 3 (STAT3) is one of the crucial transcription factors, responsible for regulating cellular proliferation, cellular differentiation, migration, programmed cell death, inflammatory response, angiogenesis, and immune activation. In this review, we have discussed the classical regulation of STAT3 via diverse growth factors, cytokines, G-protein-coupled receptors, as well as toll-like receptors. We have also highlighted the potential role of noncoding RNAs in regulating STAT3 signaling. However, the deregulation of STAT3 signaling has been found to be associated with the initiation and progression of both solid and hematological malignancies. Additionally, hyperactivation of STAT3 signaling can maintain the cancer stem cell phenotype by modulating the tumor microenvironment, cellular metabolism, and immune responses to favor drug resistance and metastasis. Finally, we have also discussed several plausible ways to target oncogenic STAT3 signaling using various small molecules derived from natural products.
Collapse
Affiliation(s)
- Manoj Garg
- Amity Institute of Molecular Medicine and Stem Cell Research (AIMMSCR), Amity University, Noida, Uttar Pradesh, India
| | - Muthu K Shanmugam
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Vipul Bhardwaj
- Amity Institute of Molecular Medicine and Stem Cell Research (AIMMSCR), Amity University, Noida, Uttar Pradesh, India
| | - Akul Goel
- La Canada High School, La Canada Flintridge, California, USA
| | - Rajat Gupta
- Amity Institute of Molecular Medicine and Stem Cell Research (AIMMSCR), Amity University, Noida, Uttar Pradesh, India
| | - Arundhiti Sharma
- Amity Institute of Molecular Medicine and Stem Cell Research (AIMMSCR), Amity University, Noida, Uttar Pradesh, India
| | - Prakash Baligar
- Amity Institute of Molecular Medicine and Stem Cell Research (AIMMSCR), Amity University, Noida, Uttar Pradesh, India
| | - Alan Prem Kumar
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Cancer Science Institute of Singapore, Center for Translational Medicine, Singapore, Singapore
| | - Boon Cher Goh
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Cancer Science Institute of Singapore, Center for Translational Medicine, Singapore, Singapore
- Department of Hematology-Oncology, National University Health System, Singapore, Singapore
| | - Lingzhi Wang
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Cancer Science Institute of Singapore, Center for Translational Medicine, Singapore, Singapore
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| |
Collapse
|
9
|
STAT3 transcription factor as target for anti-cancer therapy. Pharmacol Rep 2020; 72:1101-1124. [PMID: 32880101 DOI: 10.1007/s43440-020-00156-5] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Revised: 08/23/2020] [Accepted: 08/25/2020] [Indexed: 12/17/2022]
Abstract
STATs constitute a large family of transcription activators and transducers of signals that have an important role in many cell functions as regulation of proliferation and differentiation of the cell also regulation of apoptosis and angiogenesis. STAT3 as a member of that family, recently was discovered to have a vital role in progression of different types of cancers. The activation of STAT3 was observed to regulate multiple gene functions during cancer-like cell proliferation, differentiation, apoptosis, metastasis, inflammation, immunity, cell survival, and angiogenesis. The inhibition of STAT3 activation has been an important target for cancer therapy. Inhibitors of STAT3 have been used for a long time for treatment of many types of cancers like leukemia, melanoma, colon, and renal cancer. In this review article, we summarize and discuss different drugs inhibiting the action of STAT3 and used in treatment of different types of cancer.
Collapse
|
10
|
ApoC1 promotes the metastasis of clear cell renal cell carcinoma via activation of STAT3. Oncogene 2020; 39:6203-6217. [PMID: 32826950 DOI: 10.1038/s41388-020-01428-3] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2019] [Revised: 08/05/2020] [Accepted: 08/10/2020] [Indexed: 12/19/2022]
Abstract
Clear cell renal cell carcinoma (ccRCC) is the most common renal cancer and frequently diagnosed at an advanced stage. It is prone to develop unpredictable metastases even with proper treatment. Antiangiogenic therapy is the most effective medical treatment for metastatic ccRCC. Thus, exploration of novel approaches to inhibit angiogenesis and metastasis may potentially lead to a better therapeutic option for ccRCC. Among all the types of cancer, renal cancer samples exhibited the maximum upregulation of ApoC1 as referred to in the Oncomine database. The expression of ApoC1 was increased accompanied by ccRCC progression. A high level of ApoC1 was closely related to poor survival time in ccRCC patients. Furthermore, ApoC1 was over-expressed in the highly invasive ccRCC cells as compared to that in the low-invasive ccRCC cells. Besides, ApoC1 promoted metastasis of ccRCC cells via EMT pathway, whereas depletion of ApoC1 alleviated these effects. ApoC1 as a novel pro-metastatic factor facilitates the activation of STAT3 and enhances the metastasis of ccRCC cells. Meanwhile, ApoC1 in the exosomes were transferred from the ccRCC cells to the vascular endothelial cells and promoted metastasis of the ccRCC cells via activating STAT3. Finally, the metastatic potential of the ccRCC cells driven by ApoC1 was suppressed by DPP-4 inhibition. Our study not only identifies a novel ApoC1-STAT3 pathway in ccRCC metastasis but also provides direction for the exploration of novel strategies to predict and treat metastatic ccRCC in the future.
Collapse
|
11
|
Cerulli RA, Shehaj L, Tosic I, Jiang K, Wang J, Frank DA, Kritzer JA. Cytosolic delivery of peptidic STAT3 SH2 domain inhibitors. Bioorg Med Chem 2020; 28:115542. [PMID: 32503696 PMCID: PMC7294595 DOI: 10.1016/j.bmc.2020.115542] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Revised: 04/24/2020] [Accepted: 04/29/2020] [Indexed: 01/08/2023]
Abstract
The signal transducer and activator of transcription 3 (STAT3) protein is constitutively activated in several cancers. STAT3 activity can be blocked by inhibiting its Src Homology 2 (SH2) domain, but phosphotyrosine and its isosteres have poor bioavailability. In this work, we develop peptide-based inhibitors of STAT3-SH2 by combining chemical strategies that have proven effective for targeting other SH2 domains. These strategies include a STAT3-specific selectivity sequence, non-hydrolyzable phosphotyrosine isosteres, and a high-efficiency cell-penetrating peptide. Peptides that combined these three strategies had substantial biological stability and cytosolic delivery, as measured using highly quantitative cell-based assays. However, these peptides did not inhibit STAT3 activity in cells. By comparing in vitro binding affinity, cell penetration, and proteolytic stability, this work explores the delicate balance of factors that contribute to biological activity for peptidic inhibitors of STAT3.
Collapse
Affiliation(s)
- Robert A Cerulli
- Cell, Molecular and Developmental Biology Program, Graduate School of Biomedical Sciences, Tufts University, Boston, MA 02111, United States
| | - Livia Shehaj
- Department of Chemistry, Tufts University, Medford, MA 02155, United States
| | - Isidora Tosic
- Department of Biochemistry, Faculty of Medicine, University of Novi Sad, Novi Sad, Serbia; Dana Farber Cancer Institute, Department of Medical Oncology, Boston, MA 02215, United States
| | - Kevin Jiang
- Dana Farber Cancer Institute, Department of Medical Oncology, Boston, MA 02215, United States
| | - Jing Wang
- Department of Chemistry, Tufts University, Medford, MA 02155, United States
| | - David A Frank
- Dana Farber Cancer Institute, Department of Medical Oncology, Boston, MA 02215, United States; Brigham and Women's Hospital, Department of Medicine, Boston, MA 02115, United States; Harvard Medical School, Boston, MA 02111, United States
| | - Joshua A Kritzer
- Department of Chemistry, Tufts University, Medford, MA 02155, United States.
| |
Collapse
|
12
|
Ma JH, Qi J, Lin SQ, Zhang CY, Liu FY, Xie WD, Li X. STAT3 Targets ERR-α to Promote Epithelial-Mesenchymal Transition, Migration, and Invasion in Triple-Negative Breast Cancer Cells. Mol Cancer Res 2019; 17:2184-2195. [PMID: 31427441 DOI: 10.1158/1541-7786.mcr-18-1194] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Revised: 06/06/2019] [Accepted: 08/15/2019] [Indexed: 11/16/2022]
Abstract
STAT3 is constitutively activated in many malignant tumor types and plays an important role in multiple aspects of cancer aggressiveness. In this study, we found that estrogen-related receptor α (ERR-α) correlating with STAT3 was highly expressed in triple-negative breast cancer (TNBC) cell lines and tissues, which was associated with both the pathologic stage and prognosis of patients with TNBC. In vitro studies showed that ERR-α promoted TNBC cell migration and invasion, which was regulated by STAT3. Phosphorylated STAT3 (p-STAT3, Tyr 705) could bind to the promotor of ERR-α, and activate its transcription, which was suggested by luciferase assay and chromatin immunoprecipitation assay. We also found that ERR-α was the key target gene regulated by STAT3 in promoting epithelial-mesenchymal transition (EMT), migration, and invasion. ERR-α upregulated the expression of ZEB1, N-cadherin, and vimentin while downregulated the expression of E-cadherin. Furthermore, in vivo studies showed that ERR-α could increase the metastasis ability of TNBC. Our finding demonstrated that ERR-α was a direct regulatory gene target of p-STAT3, which was enriched for processes involving invasion and metastasis in TNBC and provided insight into TNBC pathogenesis, as well as a potential therapeutic option against TNBC metastasis. IMPLICATIONS: Our research first showed that p-STAT3 (Tyr 705) could bind to the promotor region of ERR-α and promote EMT in TNBC by ZEB1 pathways, thus providing a potential clinical target for TNBC.
Collapse
Affiliation(s)
- Jia-Hui Ma
- School of Ocean, Shandong University, Weihai, P.R. China
| | - Jie Qi
- School of Pharmaceutical Sciences, Shandong University, Jinan, P.R. China
| | - Shi-Qi Lin
- School of Ocean, Shandong University, Weihai, P.R. China
| | - Cai-Yun Zhang
- School of Ocean, Shandong University, Weihai, P.R. China
| | - Fang-Yuan Liu
- School of Ocean, Shandong University, Weihai, P.R. China
| | - Wei-Dong Xie
- School of Ocean, Shandong University, Weihai, P.R. China
| | - Xia Li
- School of Ocean, Shandong University, Weihai, P.R. China.
- School of Pharmaceutical Sciences, Shandong University, Jinan, P.R. China
- The Key Laboratory of Chemistry for Natural Product of Guizhou Province and Chinese Academy of Science, Guiyang, P.R. China
| |
Collapse
|
13
|
Liang ZT, Li J, Rong R, Wang YJ, Xiao LG, Yang GT, Zhang HQ. Ghrelin up-regulates cartilage-specific genes via the ERK/STAT3 pathway in chondrocytes of patients with adolescent idiopathic scoliosis. Biochem Biophys Res Commun 2019; 518:259-265. [PMID: 31421834 DOI: 10.1016/j.bbrc.2019.08.044] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Accepted: 08/08/2019] [Indexed: 01/19/2023]
Abstract
Adolescent idiopathic scoliosis (AIS) is a severe spinal deformity that often occurs during puberty. The occurrence of AIS is suggested to be related to abnormal development of cartilage. Our previous study found increased serum ghrelin levels in AIS patients that may linked to the development of AIS. However, whether ghrelin affects cartilage in AIS patients is unclear. We used quantitative real-time PCR (qRT-PCR) and immunohistochemistry to detect the expression of cartilage-specific genes and the ghrelin receptor, growth hormone secretagogue receptor (GHSR). The mRNA and protein levels of collagen II (COLII), SOX9, AGGRECAN (ACAN) and GHSR were higher in AIS patients than in controls. In addition, the protein levels of GHSR downstream signaling pathway members p-STAT3 (Ser727), and p-ERK1/2 were increased. Furthermore, we treated chondrocytes from AIS patients with 100 nM ghrelin, the cell proliferation assay and Western blotting showed that ghrelin promotes chondrocyte proliferation and enhances COLII, SOX9, ACAN, p-ERK1/2 and p-STAT3 expression, respectively. Interestingly, all these observed alterations were abolished by ghrelin + [D-Lys3]-GHRP-6 (a ghrelin receptor inhibitor) treatment. And after U0126 (an inhibitor of ERK1/2 phosphorylation) treatment, ERK1/2 and STAT3 (Ser727) phosphorylation was simultaneously suppressed indicating that ERK1/2 is an upstream pathway protein of STAT3 (Ser727). In conclusion, ghrelin plays an important role in upregulating cartilage-specific genes on AIS primary chondrocytes by activating ERK/STAT3 signaling pathway.
Collapse
Affiliation(s)
- Zhuo-Tao Liang
- Department of Spine Surgery, Xiangya Hospital, Central South University, No. 87, Xiangya Road, Changsha, 410008, China
| | - Jiong Li
- Department of Spine Surgery, Xiangya Hospital, Central South University, No. 87, Xiangya Road, Changsha, 410008, China
| | - Rong- Rong
- Department of Ophthalmology, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Yun-Jia Wang
- Department of Spine Surgery, Xiangya Hospital, Central South University, No. 87, Xiangya Road, Changsha, 410008, China
| | - Li-Ge Xiao
- Department of Spine Surgery, Xiangya Hospital, Central South University, No. 87, Xiangya Road, Changsha, 410008, China
| | - Guan-Teng Yang
- Department of Spine Surgery, Xiangya Hospital, Central South University, No. 87, Xiangya Road, Changsha, 410008, China
| | - Hong-Qi Zhang
- Department of Spine Surgery, Xiangya Hospital, Central South University, No. 87, Xiangya Road, Changsha, 410008, China.
| |
Collapse
|
14
|
Targeting of JAK-STAT Signaling in Breast Cancer: Therapeutic Strategies to Overcome Drug Resistance. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1152:271-281. [PMID: 31456189 DOI: 10.1007/978-3-030-20301-6_14] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Rapidly emerging ground-breaking discoveries have provided near to complete resolution of breast cancer signaling landscape and scientists have mapped the knowledge gaps associated with proteins encoded by the human genome. Based on the insights gleaned from decades of research, it seems clear that ligands transmit distinct information through specific receptors that is processed into characteristically unique biological outputs. Advances in imaging, structural biology, proteomics and genome editing have helped us to gain new insights into JAK-STAT signaling and how alterations in this pathway contributed to development of breast cancer and metastatic spread. Data obtained through high-throughput technologies has started to shed light on signal-transducer complexes formed during JAK-STAT signaling. Pharmacologists and molecular biologists are focusing on the strategies to therapeutically target this pathway to overcome drug resistance associated with breast cancer.
Collapse
|
15
|
Two decades of research in discovery of anticancer drugs targeting STAT3, how close are we? Pharmacol Ther 2018; 191:74-91. [DOI: 10.1016/j.pharmthera.2018.06.006] [Citation(s) in RCA: 113] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
16
|
Vazhappilly CG, Saleh E, Ramadan W, Menon V, Al-Azawi AM, Tarazi H, Abdu-Allah H, El-Shorbagi AN, El-Awady R. Inhibition of SHP2 by new compounds induces differential effects on RAS/RAF/ERK and PI3K/AKT pathways in different cancer cell types. Invest New Drugs 2018; 37:252-261. [PMID: 29947013 DOI: 10.1007/s10637-018-0626-5] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Accepted: 06/20/2018] [Indexed: 10/28/2022]
Abstract
Kinases and phosphatases are important players in growth signaling and are involved in cancer development. For development of targeted cancer therapy, attention is given to kinases rather than phosphatases inhibitors. Src homology region 2 domain-containing protein tyrosine phosphatase2 (SHP2) is overexpressed in different types of cancers. We investigated the SHP2-inhibitory effects of two new 5-aminosalicylate-4-thiazolinones in human cervical (HeLa) and breast (MCF-7 & MDA-MB-231) cancer cells. In-silico molecular docking showed preferential affinity of the two compounds towards the catalytic over the allosteric site of SHP2. An enzymatic assay confirmed the docking results whereby 0.01 μM of both compounds reduced SHP2 activity to 50%. On cellular level, the two compounds significantly reduced the expression of SHP2, KRAS, p-ERK and p-STAT3 in HeLa but not in the other two cell lines. Phosphorylation of AKT and JNK was enhanced in HeLa and MCF7. Both compounds exhibited anti-proliferative/anti-migratory effects on HeLa and MCF7 but not in MDA-MB-231 cells. These results indicate that inhibition of SHP2 and its downstream pathways by the two compounds might be a promising strategy for cancer therapy in some but not all cancer types.
Collapse
Affiliation(s)
- Cijo George Vazhappilly
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah, United Arab Emirates
| | - Ekram Saleh
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah, United Arab Emirates.,Cancer Biology Department, National Cancer Institute, Cairo University, Cairo, Egypt
| | - Wafaa Ramadan
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah, United Arab Emirates
| | - Varsha Menon
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah, United Arab Emirates
| | - Aya Mudhafar Al-Azawi
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah, United Arab Emirates
| | - Hamadeh Tarazi
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah, United Arab Emirates.,College of Pharmacy, University of Sharjah, University City Road, 27272, Sharjah, United Arab Emirates
| | - Hajjaj Abdu-Allah
- Medicinal Chemistry Department, College of Pharmacy, Assuit University, Assuit, Egypt
| | - Abdel-Nasser El-Shorbagi
- College of Pharmacy, University of Sharjah, University City Road, 27272, Sharjah, United Arab Emirates.,Medicinal Chemistry Department, College of Pharmacy, Assuit University, Assuit, Egypt
| | - Raafat El-Awady
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah, United Arab Emirates. .,College of Pharmacy, University of Sharjah, University City Road, 27272, Sharjah, United Arab Emirates.
| |
Collapse
|