1
|
Kciuk M, Alam M, Ali N, Rashid S, Głowacka P, Sundaraj R, Celik I, Yahya EB, Dubey A, Zerroug E, Kontek R. Epigallocatechin-3-Gallate Therapeutic Potential in Cancer: Mechanism of Action and Clinical Implications. Molecules 2023; 28:5246. [PMID: 37446908 PMCID: PMC10343677 DOI: 10.3390/molecules28135246] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 06/30/2023] [Accepted: 07/03/2023] [Indexed: 07/15/2023] Open
Abstract
Cellular signaling pathways involved in the maintenance of the equilibrium between cell proliferation and apoptosis have emerged as rational targets that can be exploited in the prevention and treatment of cancer. Epigallocatechin-3-gallate (EGCG) is the most abundant phenolic compound found in green tea. It has been shown to regulate multiple crucial cellular signaling pathways, including those mediated by EGFR, JAK-STAT, MAPKs, NF-κB, PI3K-AKT-mTOR, and others. Deregulation of the abovementioned pathways is involved in the pathophysiology of cancer. It has been demonstrated that EGCG may exert anti-proliferative, anti-inflammatory, and apoptosis-inducing effects or induce epigenetic changes. Furthermore, preclinical and clinical studies suggest that EGCG may be used in the treatment of numerous disorders, including cancer. This review aims to summarize the existing knowledge regarding the biological properties of EGCG, especially in the context of cancer treatment and prophylaxis.
Collapse
Affiliation(s)
- Mateusz Kciuk
- Department of Molecular Biotechnology and Genetics, University of Lodz, Banacha Street 12/16, 90-237 Lodz, Poland; (M.K.); (R.K.)
- Doctoral School of Exact and Natural Sciences, University of Lodz, Banacha Street 12/16, 90-237 Lodz, Poland
| | - Manzar Alam
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi 110025, India;
| | - Nemat Ali
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia;
| | - Summya Rashid
- Department of Pharmacology & Toxicology, College of Pharmacy, Prince Sattam Bin Abdulaziz University, P.O. Box 173, Al-Kharj 11942, Saudi Arabia
| | - Pola Głowacka
- Department of Medical Biochemistry, Medical University of Lodz, Mazowiecka 6/8, 90-001 Lodz, Poland;
- Doctoral School of Medical University of Lodz, Hallera 1 Square, 90-700 Lodz, Poland
| | - Rajamanikandan Sundaraj
- Department of Biochemistry, Centre for Drug Discovery, Karpagam Academy of Higher Education, Coimbatore 641021, India;
| | - Ismail Celik
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Erciyes University, Kayseri 38280, Turkey;
| | - Esam Bashir Yahya
- Bioprocess Technology Division, School of Industrial Technology, Universiti Sains Malaysia, Penang 11800, Malaysia;
| | - Amit Dubey
- Computational Chemistry and Drug Discovery Division, Quanta Calculus, Greater Noida 201310, India;
- Department of Pharmacology, Saveetha Institute of Medical and Technical Sciences, Saveetha Dental College and Hospital, Chennai 600077, India
| | - Enfale Zerroug
- LMCE Laboratory, Group of Computational and Pharmaceutical Chemistry, University of Biskra, Biskra 07000, Algeria;
| | - Renata Kontek
- Department of Molecular Biotechnology and Genetics, University of Lodz, Banacha Street 12/16, 90-237 Lodz, Poland; (M.K.); (R.K.)
| |
Collapse
|
2
|
Imran M, Insaf A, Hasan N, Sugandhi VV, Shrestha D, Paudel KR, Jha SK, Hansbro PM, Dua K, Devkota HP, Mohammed Y. Exploring the Remarkable Chemotherapeutic Potential of Polyphenolic Antioxidants in Battling Various Forms of Cancer. Molecules 2023; 28:molecules28083475. [PMID: 37110709 PMCID: PMC10142939 DOI: 10.3390/molecules28083475] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 04/10/2023] [Accepted: 04/11/2023] [Indexed: 04/29/2023] Open
Abstract
Plant-derived compounds, specifically antioxidants, have played an important role in scavenging the free radicals present under diseased conditions. The persistent generation of free radicals in the body leads to inflammation and can result in even more severe diseases such as cancer. Notably, the antioxidant potential of various plant-derived compounds prevents and deregulates the formation of radicals by initiating their decomposition. There is a vast literature demonstrating antioxidant compounds' anti-inflammatory, anti-diabetic, and anti-cancer potential. This review describes the molecular mechanism of various flavonoids, such as quercetin, kaempferol, naringenin, epicatechin, and epicatechin gallate, against different cancers. Additionally, the pharmaceutical application of these flavonoids against different cancers using nanotechnologies such as polymeric, lipid-based nanoparticles (solid-lipid and liquid-lipid), liposomes, and metallic nanocarriers is addressed. Finally, combination therapies in which these flavonoids are employed along with other anti-cancer agents are described, indicating the effective therapies for the management of various malignancies.
Collapse
Affiliation(s)
- Mohammad Imran
- Therapeutics Research Group, Frazer Institute, Faculty of Medicine, The University of Queensland, Brisbane, QLD 4102, Australia
| | - Areeba Insaf
- Department of Pharmacognosy and Phytochemistry, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Nazeer Hasan
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Vrushabh V Sugandhi
- Department of Pharmaceutics, Y.B. Chavan College of Pharmacy, Aurangabad 431001, India
| | - Deumaya Shrestha
- Department of Bioscience, Mokp o National University, Muna 58554, Republic of Korea
| | - Keshav Raj Paudel
- Centre of Inflammation, School of Life Sciences, Faculty of Science, Centenary Institute and University of Technology Sydney, Sydney, NSW 2007, Australia
| | - Saurav Kumar Jha
- Department of Biomedicine, Health & Life Convergence Sciences, Mokpo National University, Muna 58554, Republic of Korea
| | - Philip M Hansbro
- Centre of Inflammation, School of Life Sciences, Faculty of Science, Centenary Institute and University of Technology Sydney, Sydney, NSW 2007, Australia
| | - Kamal Dua
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Sydney, NSW 2007, Australia
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW 2007, Australia
| | - Hari Prasad Devkota
- Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Kumamoto 862-0973, Japan
- Pharmacy Program, Gandaki University, Pokhara 33700, Nepal
| | - Yousuf Mohammed
- Therapeutics Research Group, Frazer Institute, Faculty of Medicine, The University of Queensland, Brisbane, QLD 4102, Australia
- School of Pharmacy, The University of Queensland, Brisbane, QLD 4102, Australia
| |
Collapse
|
3
|
Mihanfar A, Yousefi B, Azizzadeh B, Majidinia M. Interactions of melatonin with various signaling pathways: implications for cancer therapy. Cancer Cell Int 2022; 22:420. [PMID: 36581900 PMCID: PMC9798601 DOI: 10.1186/s12935-022-02825-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2022] [Accepted: 12/06/2022] [Indexed: 12/30/2022] Open
Abstract
Melatonin is a neuro-hormone with conserved roles in evolution. Initially synthetized as an antioxidant molecule, it has gained prominence as a key molecule in the regulation of the circadian rhythm. Melatonin exerts its effect by binding to cytoplasmic and intra-nuclear receptors, and is able to regulate the expression of key mediators of different signaling pathways. This ability has led scholars to investigate the role of melatonin in reversing the process of carcinogenesis, a process in which many signaling pathways are involved, and regulating these pathways may be of clinical significance. In this review, the role of melatonin in regulating multiple signaling pathways with important roles in cancer progression is discussed, and evidence regarding the beneficence of targeting malignancies with this approach is presented.
Collapse
Affiliation(s)
- Ainaz Mihanfar
- grid.412763.50000 0004 0442 8645Solid Tumor Research Center, Cellular and Molecular Medicine Institute, Urmia University of Medical Sciences, Urmia, Iran
| | - Bahman Yousefi
- grid.412888.f0000 0001 2174 8913Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Bita Azizzadeh
- grid.449129.30000 0004 0611 9408Department of Biochemistry, School of Medicine, Ilam University of Medical Sciences, Ilam, Iran
| | - Maryam Majidinia
- grid.412763.50000 0004 0442 8645Solid Tumor Research Center, Cellular and Molecular Medicine Institute, Urmia University of Medical Sciences, Urmia, Iran
| |
Collapse
|
4
|
Therapeutic Effects of Green Tea Polyphenol (‒)-Epigallocatechin-3-Gallate (EGCG) in Relation to Molecular Pathways Controlling Inflammation, Oxidative Stress, and Apoptosis. Int J Mol Sci 2022; 24:ijms24010340. [PMID: 36613784 PMCID: PMC9820274 DOI: 10.3390/ijms24010340] [Citation(s) in RCA: 44] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 12/20/2022] [Accepted: 12/22/2022] [Indexed: 12/28/2022] Open
Abstract
(‒)-Epigallocatechin-3-gallate (EGCG) is the most abundant polyphenol in green tea. Thanks to multiple interactions with cell surface receptors, intracellular signaling pathways, and nuclear transcription factors, EGCG possesses a wide variety of anti-inflammatory, antioxidant, antifibrotic, anti-remodelation, and tissue-protective properties which may be useful in the treatment of various diseases, particularly in cancer, and neurological, cardiovascular, respiratory, and metabolic disorders. This article reviews current information on the biological effects of EGCG in the above-mentioned disorders in relation to molecular pathways controlling inflammation, oxidative stress, and cell apoptosis.
Collapse
|
5
|
Li XX, Liu C, Dong SL, Ou CS, Lu JL, Ye JH, Liang YR, Zheng XQ. Anticarcinogenic potentials of tea catechins. Front Nutr 2022; 9:1060783. [PMID: 36545470 PMCID: PMC9760998 DOI: 10.3389/fnut.2022.1060783] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 11/21/2022] [Indexed: 12/07/2022] Open
Abstract
Catechins are a cluster of polyphenolic bioactive components in green tea. Anticarcinogenic effects of tea catechins have been reported since the 1980s, but it has been controversial. The present paper reviews the advances in studies on the anticarcinogenic activities of tea and catechins, including epidemiological evidence and anticarcinogenic mechanism. Tea catechins showed antagonistic effects on many cancers, such as gynecological cancers, digestive tract cancers, incident glioma, liver and gallbladder cancers, lung cancer, etc. The mechanism underlying the anticarcinogenic effects of catechins involves in inhibiting the proliferation and growth of cancer cells, scavenging free radicals, suppressing metastasis of cancer cells, improving immunity, interacting with other anticancer drugs, and regulating signaling pathways. The inconsistent results and their causes are also discussed in this paper.
Collapse
Affiliation(s)
- Xiao-Xiang Li
- Tea Research Institute, Zhejiang University, Hangzhou, China
| | - Chang Liu
- Tea Science Society of China, Hangzhou, China
| | - Shu-Ling Dong
- Tea Research Institute, Zhejiang University, Hangzhou, China
| | - Can-Song Ou
- Development Center of Liubao Tea Industry, Cangwu, China
| | - Jian-Liang Lu
- Tea Research Institute, Zhejiang University, Hangzhou, China
| | - Jian-Hui Ye
- Tea Research Institute, Zhejiang University, Hangzhou, China
| | - Yue-Rong Liang
- Tea Research Institute, Zhejiang University, Hangzhou, China,*Correspondence: Yue-Rong Liang,
| | - Xin-Qiang Zheng
- Tea Research Institute, Zhejiang University, Hangzhou, China,Xin-Qiang Zheng,
| |
Collapse
|
6
|
AZ12756122, a novel fatty acid synthase inhibitor, decreases resistance features in EGFR-TKI resistant EGFR-mutated NSCLC cell models. Biomed Pharmacother 2022; 156:113942. [DOI: 10.1016/j.biopha.2022.113942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 10/25/2022] [Accepted: 10/26/2022] [Indexed: 11/22/2022] Open
|
7
|
The Role of Natural Products as Inhibitors of JAK/STAT Signaling Pathways in Glioblastoma Treatment. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:7838583. [PMID: 36193062 PMCID: PMC9526628 DOI: 10.1155/2022/7838583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 07/28/2022] [Accepted: 08/23/2022] [Indexed: 11/18/2022]
Abstract
The permeability of glioblastoma, as well as its escaping the immune system, makes them one of the most deadly human malignancies. By avoiding programmed cell death (apoptosis), unlimited cell growth and metastatic ability could dramatically affect the immune system. Genetic mutations, epigenetic changes, and overexpression of oncogenes can cause this process. On the other hand, the blood-brain barrier (BBB) and intratumor heterogeneity are important factors causing resistance to therapy. Several signaling pathways have been identified in this field, including the Janus tyrosine kinase (JAK) converter and signal transducer and activator of transcription (STAT) activator pathways, which are closely related. In addition, the JAK/STAT signaling pathway contributes to a wide array of tumorigenesis functions, including replication, anti-apoptosis, angiogenesis, and immune suppression. Introducing this pathway as the main tumorigenesis and treatment resistance center can give a better understanding of how it operates. In light of this, it is an important goal in treating many disorders, particularly cancer. The inhibition of this signaling pathway is being considered an approach to the treatment of glioblastoma. The use of natural products alternatively to conventional therapies is another area of research interest among researchers. Some natural products that originate from plants or natural sources can interfere with JAK/STAT signaling in human malignant cells, also by stopping the progression and phosphorylation of JAK/STAT, inducing apoptosis, and stopping the cell cycle. Natural products are a viable alternative to conventional chemotherapy because of their cost-effectiveness, wide availability, and almost no side effects.
Collapse
|
8
|
Zhang Z, Zhu Q, Wang S, Shi C. Epigallocatechin-3-gallate inhibits the formation of neutrophil extracellular traps and suppresses the migration and invasion of colon cancer cells by regulating STAT3/CXCL8 pathway. Mol Cell Biochem 2022; 478:887-898. [PMID: 36112238 DOI: 10.1007/s11010-022-04550-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Accepted: 08/26/2022] [Indexed: 11/30/2022]
Abstract
Colon cancer is a common malignant tumor of the digestive tract. Tea catechin exerts anti-tumor effects in colon cancer. This work aimed to determine the functions of epigallocatechin-3-gallate (EGCG), one of the main active components of Tea catechins, in the progression of colon cancer. In this work, enzyme-linked immune-sorbent assay, quantitative real-time PCR and western blotting was utilized to examine the levels of IL-1β, TNF-α, STAT3, p-STAT3 and CXCL8 in colon cancer patients and healthy controls. Compared with healthy controls, the levels of IL-1β and TNF-α were significantly increased in the peripheral blood of colon cancer patients, and the expression of STAT3, p-STAT3 and CXCL8 was elevated in the neutrophils derived from colon cancer patients. Moreover, neutrophils were treated with phorbol ester (PMA) or DNase I to induce or impede the formation of neutrophil extracellular traps (NETs). Both STAT3 overexpression and PMA treatment promoted the expression of CXCL8, myeloperoxidase (MPO) and citrullinated histone H3 (H3Cit) in the colon cancer-derived neutrophils, indicating that STAT3 overexpression facilitated the formation of NETs. STAT3 deficiency suppressed the formation of NETs, which consistent with the results of DNase I treatment. Transwell assay was utilized to detect the migration and invasion of colon cancer cell line SW480. EGCG treatment suppressed the formation of NETs and the expression of STAT3 and CXCL8 in the colon cancer-derived neutrophils, and then inhibited the migration and invasion of SW480 cells. In conclusion, this work demonstrated that EGCG inhibited the formation of NETs and subsequent suppressed the migration and invasion of colon cancer cells by regulating STAT3/CXCL8 signalling pathway. Thus, this study suggests that EGCG may become a potential drug for colon cancer therapy.
Collapse
Affiliation(s)
- Zhuoxian Zhang
- Department of Oncology, Gaoxin Branch Of The First Affiliated Hospital Of Nanchang University, No.7889, Changdong avenue, Nanchang, 330006, Jiangxi Province, People's Republic of China
| | - Qiuli Zhu
- Department of Genetics, Gaoxin Branch Of The First Affiliated Hospital Of Nanchang University, Nanchang, 330006, Jiangxi Province, People's Republic of China
| | - Siya Wang
- Department of Oncology, Gaoxin Branch Of The First Affiliated Hospital Of Nanchang University, No.7889, Changdong avenue, Nanchang, 330006, Jiangxi Province, People's Republic of China
| | - Chao Shi
- Department of Oncology, Gaoxin Branch Of The First Affiliated Hospital Of Nanchang University, No.7889, Changdong avenue, Nanchang, 330006, Jiangxi Province, People's Republic of China.
| |
Collapse
|
9
|
Umar S, Kan P, Carter MJ, Shitabata P, Novosilska M. Lichen Planopilaris Responsive to a Novel Phytoactive Botanical Treatment: A Case Series. Dermatol Ther (Heidelb) 2022; 12:1697-1710. [PMID: 35674981 PMCID: PMC9276860 DOI: 10.1007/s13555-022-00749-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 05/17/2022] [Indexed: 11/28/2022] Open
Abstract
INTRODUCTION Lichen planopilaris (LPP) is characterized by chronic scarring alopecia that is progressive and typically refractory to therapy. Current drug treatments are suboptimal and not applicable for long-term use because of the high potential for adverse effects, warranting safer and more effective treatment alternatives. METHODS Based on our previous success in treating a patient with central centrifugal cicatricial alopecia using a topical botanical formulation (Gashee), we reviewed records of four patients with biopsy-proven LPP treated with the topical formulation alone or in combination with its oral preparation. Three patients had failed previous treatment with intralesional steroid injections, topical minoxidil, tacrolimus, and clobetasol. Physical examination and photographic documentation were also used as outcome measures. Treatment duration with the botanical formulations ranged from 6 weeks to 9.5 months. RESULTS All patients showed overall improvement in surrogate indicators of LPP activity as evidenced by the disappearance of symptoms (pruritus, tenderness, scalp irritation, and hair shedding), improvement in hair growth, and reduction in redness. All reported a high satisfaction level and no adverse effects. CONCLUSIONS Patients with treatment-refractory LPP responded to a novel botanical treatment. To the best of our knowledge, this is the first published report of LPP responding to a plant-based natural treatment. Further evaluation of this treatment in a controlled trial with a larger number of patients is warranted.
Collapse
Affiliation(s)
- Sanusi Umar
- Department of Medicine, University of California at Los Angeles, Los Angeles, CA, USA. .,Division of Dermatology, Department of Medicine, Harbor-UCLA Medical Center, Torrance, CA, USA. .,Dr. U Hair and Skin Clinic, 2121 N. Sepulveda Avenue, Suite 200, Manhattan Beach, CA, 90266, USA.
| | - Petrina Kan
- Department of Molecular Biology, University of California at Los Angeles, Los Angeles, CA, USA
| | | | - Paul Shitabata
- Department of Medicine, University of California at Los Angeles, Los Angeles, CA, USA.,Division of Dermatology, Department of Medicine, Harbor-UCLA Medical Center, Torrance, CA, USA.,Dermatopathology Institute, Torrance, CA, USA
| | - Myroslava Novosilska
- Department of Dermatology and Oncology, Aesthetic Medical Clinic Myroslava Novosilska, Lviv, Ukraine
| |
Collapse
|
10
|
Sahadevan R, Singh S, Binoy A, Sadhukhan S. Chemico-biological aspects of (-)-epigallocatechin- 3-gallate (EGCG) to improve its stability, bioavailability and membrane permeability: Current status and future prospects. Crit Rev Food Sci Nutr 2022; 63:10382-10411. [PMID: 35491671 DOI: 10.1080/10408398.2022.2068500] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Natural products have been a bedrock for drug discovery for decades. (-)-Epigallocatechin-3-gallate (EGCG) is one of the widely studied natural polyphenolic compounds derived from green tea. It is the key component believed to be responsible for the medicinal value of green tea. Significant studies implemented in in vitro, in cellulo, and in vivo models have suggested its anti-oxidant, anti-cancer, anti-diabetic, anti-inflammatory, anti-microbial, neuroprotective activities etc. Despite having such a wide array of therapeutic potential and promising results in preclinical studies, its applicability to humans has encountered with rather limited success largely due to the poor bioavailability, poor membrane permeability, rapid metabolic clearance and lack of stability of EGCG. Therefore, novel techniques are warranted to address those limitations so that EGCG or its modified analogs can be used in the clinical setup. This review comprehensively covers different strategies such as structural modifications, nano-carriers as efficient drug delivery systems, synergistic studies with other bioactivities to improve the chemico-biological aspects (e.g., stability, bioavailability, permeability, etc.) of EGCG for its enhanced pharmacokinetics and pharmacological properties, eventually enhancing its therapeutic potentials. We think this review article will serve as a strong platform with comprehensive literature on the development of novel techniques to improve the bioavailability of EGCG so that it can be translated to the clinical applications.
Collapse
Affiliation(s)
- Revathy Sahadevan
- Department of Chemistry, Indian Institute of Technology Palakkad, Kerala, India
| | - Satyam Singh
- Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Madhya Pradesh, India
| | - Anupama Binoy
- Department of Chemistry, Indian Institute of Technology Palakkad, Kerala, India
| | - Sushabhan Sadhukhan
- Department of Chemistry, Indian Institute of Technology Palakkad, Kerala, India
- Department of Biological Sciences and Engineering, Indian Institute of Technology Palakkad, Kerala, India
| |
Collapse
|
11
|
Yu W, Ma Y, Shrivastava SK, Srivastava RK, Shankar S. Chronic alcohol exposure induces hepatocyte damage by inducing oxidative stress, SATB2 and stem cell‐like characteristics, and activating lipogenesis. J Cell Mol Med 2022; 26:2119-2131. [PMID: 35152538 PMCID: PMC8980954 DOI: 10.1111/jcmm.17235] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 01/25/2022] [Accepted: 01/31/2022] [Indexed: 12/12/2022] Open
Abstract
Alcohol is a risk factor for hepatocellular carcinoma (HCC). However, the molecular mechanism by which chronic alcohol consumption contributes to HCC is not well understood. The purpose of the study was to demonstrate the effects of chronic ethanol exposure on the damage of human normal hepatocytes. Our data showed that chronic exposure of hepatocytes with ethanol induced changes similar to transformed hepatocytes that is, exhibited colonies and anchorage‐independent growth. These damaged hepatocytes contained high levels of reactive oxygen species (ROS) and showed induction of the SATB2 gene. Furthermore, damaged hepatocytes gained the phenotypes of CSCs which expressed stem cell markers (CD133, CD44, CD90, EpCAM, AFP and LGR5), and pluripotency maintaining factors (Sox‐2, POU5F1/Oct4 and KLF‐4). Ethanol exposure also induced Nanog, a pluripotency maintaining transcription factor that functions in concert with Oct4 and SOX‐2. Furthermore, ethanol induced expression of EMT‐related transcription factors (Snail, Slug and Zeb1), N‐Cadherin, and inhibited E‐cadherin expression in damaged hepatocytes. Ethanol enhanced recruitment of SATB2 to promoters of Bcl‐2, Nanog, c‐Myc, Klf4 and Oct4. Ethanol also induced activation of the Wnt/TCF‐LEF1 pathway and its targets (Bcl‐2, Cyclin D1, AXIN2 and Myc). Finally, ethanol induced hepatocellular steatosis, SREBP1 transcription, and modulated the expression of SREBP1c, ACAC, ACLY, FASN, IL‐1β, IL‐6, TNF‐α, GPC3, FLNB and p53. These data suggest that chronic alcohol consumption may contribute towards the development of HCC by damaging normal hepatocytes with the generation of inflammatory environment, induction of SATB2, stem cell‐like characteristics, and cellular steatosis.
Collapse
Affiliation(s)
- Wei Yu
- Kansas City VA Medical Center Kansas City Missouri USA
| | - Yiming Ma
- Kansas City VA Medical Center Kansas City Missouri USA
| | - Sushant K. Shrivastava
- Department of Pharmaceutics Indian Institute of Technology Banaras Hindu University Varanasi U.P. India
| | - Rakesh K. Srivastava
- Kansas City VA Medical Center Kansas City Missouri USA
- Department of Genetics Louisiana State University Health Sciences Center New Orleans Louisina USA
- Stanley S. Scott Cancer Center Department of Genetics Louisiana State University Health Sciences Center New Orleans Louisina USA
- A.B. Freeman School of Business Tulane University New Orleans Louisina USA
| | - Sharmila Shankar
- Kansas City VA Medical Center Kansas City Missouri USA
- John W. Deming Department of Medicine Tulane University School of Medicine New Orleans Louisina USA
- Southeast Louisiana Veterans Health Care System New Orleans Louisina USA
| |
Collapse
|
12
|
Ichikawa T, Sugamoto K, Matsuura Y, Kunitake H, Shimoda K, Morishita K. Inhibition of ATL cell proliferation by polymerized proanthocyanidin from blueberry leaves through JAK proteolysis. Cancer Sci 2022; 113:1406-1416. [PMID: 35100463 PMCID: PMC8990289 DOI: 10.1111/cas.15277] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 12/27/2021] [Accepted: 01/19/2022] [Indexed: 11/29/2022] Open
Abstract
We have previously reported that the proanthocyanidin (PAC) fraction of blueberry leaf extract (BB-PAC) inhibits the proliferation of human T lymphotropic retrovirus (HTLV-1)-infected adult T-cell leukemia (ATL) by inducing apoptosis. In the present study, we further analyzed the structure of BB-PAC and elucidated the molecular mechanism underlying the inhibitory function of HTLV-1 infected and ATL cells. After hot water extraction with fractionation with methanol-acetone, BB-PAC was found to be concentrated in fractions 4 to 7 (Fr 7). The strongest inhibition of ATL cell growth was observed with Fr7, which contained the highest BB-PAC polymerization degree of 14. The basic structure of BB-PAC is mainly B-type bonds, with A-type bonds (7.1%) and cinchonain I units as the terminal unit (6.1%). The molecular mechanism of cytotoxicity observed around Fr7 against ATL cells was the degradation of JAK1 to 3 and the dephosphorylation of STAT3/5, which occurs by proteasome-dependent proteolysis, confirming that PAC directly binds to HSP90. JAK degradation was caused by proteasome-dependent proteolysis, and we identified the direct binding of PAC to HSP90. In addition, the binding of cochaperone ATPase homolog 1 (AHA1) to HSP90, which is required for activation of the cofactor HSP90, was inhibited by BB-PAC treatment. Therefore, BB-PAC inhibited the formation of the HSP90/AHA1 complex and promoted the degradation of JAK protein due to HSP90 dysfunction. These results suggest that the highly polymerized PAC component from blueberry leaves has great potential as a preventive and therapeutic agent against HTLV-1 infected and ATL cells.
Collapse
Affiliation(s)
- Tomonaga Ichikawa
- Division of Tumor and Cellular Biochemistry, Department of Medical Sciences, University of Miyazaki, Miyazaki, Japan
| | - Kazuhiro Sugamoto
- Department of Applied Chemistry, Faculty of Engineering, University of Miyazaki, Miyazaki, Japan
| | - Yasushi Matsuura
- Miyazaki Prefectural Food Research and Development Center, Miyazaki, Japan
| | - Hisato Kunitake
- Department of Biochemistry and Applied Biosciences, Faculty of Agriculture, University of Miyazaki, Miyazaki, Japan.,Director of Center for Collaborative Research & Community Cooperation, University of Miyazaki, Japan
| | - Kazuya Shimoda
- Division of Hematology, Diabetes, and Endocrinology, Department of Internal Medicine, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan
| | - Kazuhiro Morishita
- Division of Tumor and Cellular Biochemistry, Department of Medical Sciences, University of Miyazaki, Miyazaki, Japan.,Project for Advanced Medical Research and Development, Project Research Division, Frontier Science Research Center, University of Miyazaki, Miyazaki, Japan
| |
Collapse
|
13
|
Jit BP, Pradhan B, Dash R, Bhuyan PP, Behera C, Behera RK, Sharma A, Alcaraz M, Jena M. Phytochemicals: Potential Therapeutic Modulators of Radiation Induced Signaling Pathways. Antioxidants (Basel) 2021; 11:antiox11010049. [PMID: 35052553 PMCID: PMC8773162 DOI: 10.3390/antiox11010049] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 12/22/2021] [Accepted: 12/24/2021] [Indexed: 02/06/2023] Open
Abstract
Ionizing radiation results in extensive damage to biological systems. The massive amount of ionizing radiation from nuclear accidents, radiation therapy (RT), space exploration, and the nuclear battlefield leads to damage to biological systems. Radiation injuries, such as inflammation, fibrosis, and atrophy, are characterized by genomic instability, apoptosis, necrosis, and oncogenic transformation, mediated by the activation or inhibition of specific signaling pathways. Exposure of tumors or normal cells to different doses of ionizing radiation could lead to the generation of free radical species, which can release signal mediators and lead to harmful effects. Although previous FDA-approved agents effectively mitigate radiation-associated toxicities, their use is limited due to their high cellular toxicities. Preclinical and clinical findings reveal that phytochemicals derived from plants that exhibit potent antioxidant activities efficiently target several signaling pathways. This review examined the prospective roles played by some phytochemicals in altering signal pathways associated with radiation response.
Collapse
Affiliation(s)
- Bimal Prasad Jit
- School of Life Sciences, Sambalpur University, Jyoti Vihar, Burla 768019, India; (B.P.J.); (R.D.); (R.K.B.)
- Department of Biochemistry, AIIMS, Ansari Nagar, New Delhi 110029, India;
| | - Biswajita Pradhan
- Algal Biotechnology and Molecular Systematic Laboratory, Post Graduate Department of Botany, Berhampur University, Bhanja Bihar, Berhampur 760007, India; (B.P.); (C.B.)
- Department of Biotechnology, Sangmyung University, Seoul 03016, Korea
| | - Rutumbara Dash
- School of Life Sciences, Sambalpur University, Jyoti Vihar, Burla 768019, India; (B.P.J.); (R.D.); (R.K.B.)
| | - Prajna Paramita Bhuyan
- Department of Botany, Maharaja Sriram Chandra Bhanja Deo University, Baripada 757003, India;
| | - Chhandashree Behera
- Algal Biotechnology and Molecular Systematic Laboratory, Post Graduate Department of Botany, Berhampur University, Bhanja Bihar, Berhampur 760007, India; (B.P.); (C.B.)
| | - Rajendra Kumar Behera
- School of Life Sciences, Sambalpur University, Jyoti Vihar, Burla 768019, India; (B.P.J.); (R.D.); (R.K.B.)
| | - Ashok Sharma
- Department of Biochemistry, AIIMS, Ansari Nagar, New Delhi 110029, India;
| | - Miguel Alcaraz
- Radiology and Physical Medicine Department, School of Medicine, Campus de Excelencia Internacional de Ámbito Regional (CEIR)-Campus Mare Nostrum (CMN), Universidad de Murcia, 30100 Murcia, Spain
- Correspondence: (M.A.); (M.J.); Tel.: +34-868883601 (M.A.); +91-7978478950 (M.J.)
| | - Mrutyunjay Jena
- Algal Biotechnology and Molecular Systematic Laboratory, Post Graduate Department of Botany, Berhampur University, Bhanja Bihar, Berhampur 760007, India; (B.P.); (C.B.)
- Correspondence: (M.A.); (M.J.); Tel.: +34-868883601 (M.A.); +91-7978478950 (M.J.)
| |
Collapse
|
14
|
Kubczak M, Szustka A, Rogalińska M. Molecular Targets of Natural Compounds with Anti-Cancer Properties. Int J Mol Sci 2021; 22:ijms222413659. [PMID: 34948455 PMCID: PMC8708931 DOI: 10.3390/ijms222413659] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 12/13/2021] [Accepted: 12/14/2021] [Indexed: 12/13/2022] Open
Abstract
Cancer is the second leading cause of death in humans. Despite rapid developments in diagnostic methods and therapies, metastasis and resistance to administrated drugs are the main obstacles to successful treatment. Therefore, the main challenge should be the diagnosis and design of optimal therapeutic strategies for patients to increase their chances of responding positively to treatment and increase their life expectancy. In many types of cancer, a deregulation of multiple pathways has been found. This includes disturbances in cellular metabolism, cell cycle, apoptosis, angiogenesis, or epigenetic modifications. Additionally, signals received from the microenvironment may significantly contribute to cancer development. Chemical agents obtained from natural sources seem to be very attractive alternatives to synthetic compounds. They can exhibit similar anti-cancer potential, usually with reduced side effects. It was reported that natural compounds obtained from fruits and vegetables, e.g., polyphenols, flavonoids, stilbenes, carotenoids and acetogenins, might be effective against cancer cells in vitro and in vivo. Several published results indicate the activity of natural compounds on protein expression by its influence on transcription factors. They could also be involved in alterations in cellular response, cell signaling and epigenetic modifications. Such natural components could be used in our diet for anti-cancer protection. In this review, the activities of natural compounds, including anti-cancer properties, are described. The influence of natural agents on cancer cell metabolism, proliferation, signal transduction and epigenetic modifications is highlighted.
Collapse
Affiliation(s)
- Małgorzata Kubczak
- Department of General Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/143, 90-237 Łódź, Poland;
- Department of Cytobiochemistry, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/143, 90-237 Łódź, Poland;
| | - Aleksandra Szustka
- Department of Cytobiochemistry, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/143, 90-237 Łódź, Poland;
| | - Małgorzata Rogalińska
- Department of Cytobiochemistry, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/143, 90-237 Łódź, Poland;
- Correspondence:
| |
Collapse
|
15
|
Epithelial-to-Mesenchymal Transition Is Not a Major Modulating Factor in the Cytotoxic Response to Natural Products in Cancer Cell Lines. Molecules 2021; 26:molecules26195858. [PMID: 34641401 PMCID: PMC8512490 DOI: 10.3390/molecules26195858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 09/20/2021] [Accepted: 09/22/2021] [Indexed: 12/24/2022] Open
Abstract
Numerous natural products exhibit antiproliferative activity against cancer cells by modulating various biological pathways. In this study, we investigated the potential use of eight natural compounds (apigenin, curcumin, epigallocatechin gallate, fisetin, forskolin, procyanidin B2, resveratrol, urolithin A) and two repurposed agents (fulvestrant and metformin) as chemotherapy enhancers and mesenchymal-to-epithelial (MET) inducers of cancer cells. Screening of these compounds in various colon, breast, and pancreatic cancer cell lines revealed anti-cancer activity for all compounds, with curcumin being the most effective among these in all cell lines. Although some of the natural products were able to induce MET in some cancer cell lines, the MET induction was not related to increased synergy with either 5-FU, irinotecan, gemcitabine, or gefitinib. When synergy was observed, for example with curcumin and irinotecan, this was unrelated to MET induction, as assessed by changes in E-cadherin and vimentin expression. Our results show that MET induction is compound and cell line specific, and that MET is not necessarily related to enhanced chemosensitivity.
Collapse
|
16
|
Effect of (-)-Epigallocatechin Gallate on Activation of JAK/STAT Signaling Pathway by Staphylococcal Enterotoxin A. Toxins (Basel) 2021; 13:toxins13090609. [PMID: 34564613 PMCID: PMC8473440 DOI: 10.3390/toxins13090609] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Revised: 08/22/2021] [Accepted: 08/25/2021] [Indexed: 11/17/2022] Open
Abstract
Staphylococcal enterotoxin A (SEA), which is a superantigen toxin protein, binds to cytokine receptor gp130. Gp130 activates intracellular signaling pathways, including the Janus kinase/signal transducers and activators of transcription (JAK/STAT) pathway. The effects of SEA on the JAK/STAT signaling pathway in mouse spleen cells were examined. After treatment with SEA, mRNA expression levels of interferon gamma (IFN-γ) and suppressor of cytokine-signaling 1 (SOCS1) increased. SEA-induced IFN-γ and SOCS1 expression were decreased by treatment with (-)-epigallocatechin gallate (EGCG). The phosphorylated STAT3, Tyr705, increased significantly in a SEA concentration-dependent manner in mouse spleen cells. Although (-)-3″-Me-EGCG did not inhibit SEA-induced phosphorylated STAT3, EGCG and (-)-4″-Me-EGCG significantly inhibited SEA-induced phosphorylated STAT3. It was thought that the hydroxyl group at position 3 of the galloyl group in the EGCG was responsible for binding to SEA and suppressing SEA-induced phosphorylation of STAT3. Through protein thermal shift assay in vitro, the binding of the gp130 receptor to SEA and the phosphorylation of STAT3 were inhibited by the interaction between EGCG and SEA. As far as we know, this is the first report to document that EGCG inhibits the binding of the gp130 receptor to SEA and the associated phosphorylation of STAT3.
Collapse
|
17
|
Shah D, Gandhi M, Kumar A, Cruz-Martins N, Sharma R, Nair S. Current insights into epigenetics, noncoding RNA interactome and clinical pharmacokinetics of dietary polyphenols in cancer chemoprevention. Crit Rev Food Sci Nutr 2021; 63:1755-1791. [PMID: 34433338 DOI: 10.1080/10408398.2021.1968786] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Several studies have reported the health-beneficial effects of dietary phytochemicals, namely polyphenols, to prevent various diseases, including cancer. Polyphenols, like (-)-epigallocatechin-3-gallate (EGCG) from green tea, curcumin from turmeric, and ellagic acid from pomegranate are known to act by modulating antioxidant, anti-inflammatory and apoptotic signal transduction pathways in the tumor milieu. The evolving literature underscores the role of epigenetic regulation of genes associated with cancer by these polyphenols, primarily via non-coding RNAs (ncRNAs), such as microRNAs (miRNA) and long noncoding RNA (lncRNA). However, there is little clarity on the exact role(s) played by these ncRNAs and their interactions with other ncRNAs, or with their protein targets, in response to modulation by these dietary polyphenols. Here, we review ncRNA interactions and functional networks of the complex ncRNA interactome with their targets in preclinical studies along with the role of epigenetics as well as key aspects of pharmacokinetics and phytochemistry of dietary polyphenols. We also summarize the current state of clinical trials with these dietary polyphenols. Taken together, this synthetic review provides insights into the molecular aspects underlying the anticancer chemopreventive effects of dietary polyphenols as well as summarizes data on novel biomarkers modulated by these polyphenols for preventive or therapeutic purposes in various types of cancer.
Collapse
Affiliation(s)
| | | | - Arun Kumar
- Division of Radiation Biosciences, Institute of Nuclear Medicine and Allied Sciences, Defence Research and Development Organization, Timarpur Delhi, India
| | - Natália Cruz-Martins
- Faculty of Medicine, University of Porto, Porto, Portugal.,Institute for research and Innovation in Health (i3S), University of Porto, Porto, Portugal.,Institute of Research and Advanced Training in Health Sciences and Technologies (CESPU), Gandra PRD, Portugal
| | - Rohit Sharma
- Department of Rasa Shastra and Bhaishajya Kalpana, Faculty of Ayurveda, Institute of Medical Sciences, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | | |
Collapse
|
18
|
Karami E, Esfahrood ZR, Mansouri R, Haerian A, Abdian-Asl A. Effect of epigallocatechin-3-gallate on tumor necrosis factor-alpha production by human gingival fibroblasts stimulated with bacterial lipopolysaccharide: An in vitro study. J Indian Soc Periodontol 2021; 25:11-16. [PMID: 33642735 PMCID: PMC7904023 DOI: 10.4103/jisp.jisp_323_20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 09/24/2020] [Accepted: 10/11/2020] [Indexed: 11/26/2022] Open
Abstract
Background: Evidence shows that epigallocatechin-3-gallate (EGCG) in green tea has anti-inflammatory effects. Aim: This study assessed the effect of EGCG on the production of tumor necrosis factor-alpha (TNF-α) as an inflammatory cytokine in periodontitis, which produced by human gingival fibroblasts (HGFs) stimulated with lipopolysaccharide (LPS) of Porphyromonas gingivalis. Materials and Methods: In this study, HGFs were cultured and subjected to LPS and EGCG. Cell viability of different concentrations of EGCG (10, 25, 50, 75, and 100 μM) and LPS (1, 10, 20, and 50 μg/mL) was assessed using methyl-thiazole-tetrazolium (MTT) assay. Then, the best concentrations of EGCG and P. gingivalis LPS were used simultaneously and separately to assess the production of TNF-α by HGFs using the enzyme-linked immunosorbent assay (ELISA). Assessments were done at 1, 3, and 5 days. Data were read using the ELISA reader and analyzed by the SPSS through two-way ANOVA. Results: LPS at 1, 10, and 20 and EGCG at 10.25 and 50 μM showed the least cytotoxicity in MTT assay. ELISA showed EGCG alone decreased the production of TNF-α in all days, except 10 μM on day 1. 1, 10, and 20 μg/mL LPS increased the output of TNF-α on days 1 and 3 while reducing it on day 5. The combination of EGCG and LPS showed a decrease of TNF-α in all days except on day 5 that revealed an increase in the production of TNF-α at 25 and 50 μM EGCG. Conclusion: In the combination use of EGCG and LPS, EGCG shows anti-inflammatory effects by decreasing the production of TNF-α by HGFs stimulated with P. gingivalis.
Collapse
Affiliation(s)
- Elahe Karami
- Department of Periodontics, School of Dentistry, Shahid Sadughi University of Medical Science, Yazd, Iran
| | - Zeinab Rezaei Esfahrood
- Department of Periodontics, School of Dentistry, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Reza Mansouri
- Department of Immunology, Shahid Sadoughi University of Medical Sciences, Yazd, Iran.,Blood and oncology research center, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Ahmad Haerian
- Department of Periodontics, School of Dentistry, Shahid Sadughi University of Medical Sciences, Yazd, Iran
| | - Amir Abdian-Asl
- Department of Immunology, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| |
Collapse
|
19
|
Aziz MA, Sarwar MS, Akter T, Uddin MS, Xun S, Zhu Y, Islam MS, Hongjie Z. Polyphenolic molecules targeting STAT3 pathway for the treatment of cancer. Life Sci 2021; 268:118999. [PMID: 33421525 DOI: 10.1016/j.lfs.2020.118999] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 12/22/2020] [Accepted: 12/24/2020] [Indexed: 01/17/2023]
Abstract
Cancer is accounted as the second-highest cause of morbidity and mortality throughout the world. Numerous preclinical and clinical investigations have consistently highlighted the role of natural polyphenolic compounds against various cancers. A plethora of potential bioactive polyphenolic molecules, primarily flavonoids, phenolic acids, lignans and stilbenes, have been explored from the natural sources for their chemopreventive and chemoprotective activities. Moreover, combinations of these polyphenols with current chemotherapeutic agents have also demonstrated their strong role against both progression and resistance of malignancies. Signal transducer and activator of transcription 3 (STAT3) is a ubiquitously-expressed signaling molecule in almost all body cells. Thousands of literatures have revealed that STAT3 plays significant roles in promoting the cellular proliferation, differentiation, cell cycle progression, metastasis, angiogenesis and immunosuppression as well as chemoresistance through the regulation of its downstream target genes such as Bcl-2, Bcl-xL, cyclin D1, c-Myc and survivin. For its key role in cancer development, researchers considered STAT3 as a major target for cancer therapy that mainly focuses on abrogating the expression (activation or phosphorylation) of STAT3 in tumor cells both directly and indirectly. Polyphenolic molecules have explicated their protective actions in malignant cells via targeting STAT3 both in vitro and in vivo. In this article, we reviewed how polyphenolic compounds as well as their combinations with other chemotherapeutic drugs inhibit cancer cells by targeting STAT3 signaling pathway.
Collapse
Affiliation(s)
- Md Abdul Aziz
- Department of Pharmacy, Faculty of Science, Noakhali Science and Technology University, Noakhali 3814, Bangladesh
| | - Md Shahid Sarwar
- Department of Pharmacy, Faculty of Science, Noakhali Science and Technology University, Noakhali 3814, Bangladesh.
| | - Tahmina Akter
- Department of Pharmacy, Faculty of Science, Noakhali Science and Technology University, Noakhali 3814, Bangladesh
| | - Md Sahab Uddin
- Department of Pharmacy, Southeast University, Dhaka, Bangladesh; Pharmakon Neuroscience Research Network, Dhaka, Bangladesh
| | - Song Xun
- School of Pharmaceutical Science, Health Science Center, Shenzhen University, Shenzhen, China
| | - Yu Zhu
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, SAR, China
| | - Mohammad Safiqul Islam
- Department of Pharmacy, Faculty of Science, Noakhali Science and Technology University, Noakhali 3814, Bangladesh
| | - Zhang Hongjie
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, SAR, China.
| |
Collapse
|
20
|
Wang K, Zhang FL, Jia W. Glutathione S‑transferase ω 1 promotes the proliferation, migration and invasion, and inhibits the apoptosis of non‑small cell lung cancer cells, via the JAK/STAT3 signaling pathway. Mol Med Rep 2021; 23:71. [PMID: 33236161 PMCID: PMC7716429 DOI: 10.3892/mmr.2020.11709] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 10/21/2020] [Indexed: 12/25/2022] Open
Abstract
Glutathione S‑transferase ω 1 (GSTO1) expression levels have been discovered to be upregulated in various types of cancer. However, to the best of our knowledge, the role of GSTO1 in non‑small cell lung cancer (NSCLC) has not been investigated. The present study aimed to investigate the role of GSTO1 in NSCLC and to determine the potential molecular mechanism. GSTO1 expression levels in A549 cells were knocked down using short hairpin RNA and GSTO1 overexpression in H2122 cells was achieved using cDNA constructs. Reverse transcription‑quantitative PCR was used to analyze the mRNA expression levels of GSTO1. Cell proliferation was determined using a Cell Counting Kit‑8 assay, whereas cell migration and invasion were analyzed using Transwell assays. Flow cytometric analysis was performed to determine the levels of cell apoptosis. The expression levels of GSTO1, Bax, caspase 3, JAK and STAT3 were analyzed using western blotting. The results revealed that GSTO1 overexpression significantly promoted the proliferation, migration and invasion, and inhibited the apoptosis of H2122 cells, whereas the opposite trend was achieved in A549 cells with GSTO1 knockdown. GSTO1 overexpression also significantly increased the phosphorylation levels of JAK and STAT3, whereas the knockdown of GSTO1 promoted the opposite effects. In conclusion, the findings of the present study indicated that GSTO1 may serve as an oncogene in NSCLC. The results suggested that GSTO1 may have an important role in NSCLC by regulating the JAK/STAT3 signaling pathway. Therefore, inhibiting the expression levels of GSTO1 may represent a potential novel therapeutic strategy for NSCLC.
Collapse
Affiliation(s)
- Kai Wang
- Department of Respiratory and Critical Care Medicine, Tianjin Chest Hospital, Tianjin 300222, P.R. China
| | - Fu-Lian Zhang
- Integrated TCM and Western Medicine Department, Tianjin Medical University Chu Hsien-I Memorial Hospital, Tianjin 300134, P.R. China
| | - Wei Jia
- Department of Respiratory and Critical Care Medicine, Tianjin Chest Hospital, Tianjin 300222, P.R. China
| |
Collapse
|
21
|
Caston RA, Shah F, Starcher CL, Wireman R, Babb O, Grimard M, McGeown J, Armstrong L, Tong Y, Pili R, Rupert J, Zimmers TA, Elmi AN, Pollok KE, Motea EA, Kelley MR, Fishel ML. Combined inhibition of Ref-1 and STAT3 leads to synergistic tumour inhibition in multiple cancers using 3D and in vivo tumour co-culture models. J Cell Mol Med 2021; 25:784-800. [PMID: 33274592 PMCID: PMC7812272 DOI: 10.1111/jcmm.16132] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 10/19/2020] [Accepted: 11/09/2020] [Indexed: 12/26/2022] Open
Abstract
With a plethora of molecularly targeted agents under investigation in cancer, a clear need exists to understand which pathways can be targeted simultaneously with multiple agents to elicit a maximal killing effect on the tumour. Combination therapy provides the most promise in difficult to treat cancers such as pancreatic. Ref-1 is a multifunctional protein with a role in redox signalling that activates transcription factors such as NF-κB, AP-1, HIF-1α and STAT3. Formerly, we have demonstrated that dual targeting of Ref-1 (redox factor-1) and STAT3 is synergistic and decreases cell viability in pancreatic cancer cells. Data presented here extensively expands upon this work and provides further insights into the relationship of STAT3 and Ref-1 in multiple cancer types. Using targeted small molecule inhibitors, Ref-1 redox signalling was blocked along with STAT3 activation, and tumour growth evaluated in the presence and absence of the relevant tumour microenvironment. Our study utilized qPCR, cytotoxicity and in vivo analysis of tumour and cancer-associated fibroblasts (CAF) response to determine the synergy of Ref-1 and STAT3 inhibitors. Overall, pancreatic tumours grown in the presence of CAFs were sensitized to the combination of STAT3 and Ref-1 inhibition in vivo. In vitro bladder and pancreatic cancer demonstrated the most synergistic responses. By disabling both of these important pathways, this combination therapy has the capacity to hinder crosstalk between the tumour and its microenvironment, leading to improved tumour response.
Collapse
Affiliation(s)
- Rachel A. Caston
- Department of Pediatrics and Herman B Wells Center for Pediatric ResearchIndiana University School of MedicineIndianapolisINUSA
| | - Fenil Shah
- Department of Pediatrics and Herman B Wells Center for Pediatric ResearchIndiana University School of MedicineIndianapolisINUSA
| | - Colton L. Starcher
- Department of Biochemistry and Molecular BiologyIndiana University School of MedicineIndianapolisINUSA
| | - Randall Wireman
- Department of Pediatrics and Herman B Wells Center for Pediatric ResearchIndiana University School of MedicineIndianapolisINUSA
| | - Olivia Babb
- Department of Pediatrics and Herman B Wells Center for Pediatric ResearchIndiana University School of MedicineIndianapolisINUSA
| | - Michelle Grimard
- Department of Pediatrics and Herman B Wells Center for Pediatric ResearchIndiana University School of MedicineIndianapolisINUSA
| | - Jack McGeown
- Department of Pediatrics and Herman B Wells Center for Pediatric ResearchIndiana University School of MedicineIndianapolisINUSA
| | - Lee Armstrong
- Department of Pediatrics and Herman B Wells Center for Pediatric ResearchIndiana University School of MedicineIndianapolisINUSA
| | - Yan Tong
- Department of BiostatisticsIndiana University School of MedicineIndianapolisINUSA
| | - Roberto Pili
- Department of Pharmacology and ToxicologyIndiana University School of MedicineIndianapolisINUSA
- Department of UrologyIndiana University School of MedicineIndianapolisINUSA
- Department of Hematology and OncologyIndiana University School of MedicineIndianapolisINUSA
- Indiana University Simon Comprehensive Cancer CenterIndiana University School of MedicineIndianapolisINUSA
| | - Joseph Rupert
- Department of Biochemistry and Molecular BiologyIndiana University School of MedicineIndianapolisINUSA
- Indiana University Simon Comprehensive Cancer CenterIndiana University School of MedicineIndianapolisINUSA
- Department of SurgeryIndiana University School of MedicineIndianapolisINUSA
| | - Teresa A. Zimmers
- Department of Biochemistry and Molecular BiologyIndiana University School of MedicineIndianapolisINUSA
- Indiana University Simon Comprehensive Cancer CenterIndiana University School of MedicineIndianapolisINUSA
- Department of SurgeryIndiana University School of MedicineIndianapolisINUSA
- Richard L. Roudebush Veterans Administration Medical CenterIndianapolisINUSA
| | - Adily N. Elmi
- Department of Pediatrics and Herman B Wells Center for Pediatric ResearchIndiana University School of MedicineIndianapolisINUSA
| | - Karen E. Pollok
- Department of Pediatrics and Herman B Wells Center for Pediatric ResearchIndiana University School of MedicineIndianapolisINUSA
- Department of Pharmacology and ToxicologyIndiana University School of MedicineIndianapolisINUSA
- Indiana University Simon Comprehensive Cancer CenterIndiana University School of MedicineIndianapolisINUSA
| | - Edward A. Motea
- Department of Biochemistry and Molecular BiologyIndiana University School of MedicineIndianapolisINUSA
- Indiana University Simon Comprehensive Cancer CenterIndiana University School of MedicineIndianapolisINUSA
| | - Mark R. Kelley
- Department of Pediatrics and Herman B Wells Center for Pediatric ResearchIndiana University School of MedicineIndianapolisINUSA
- Department of Biochemistry and Molecular BiologyIndiana University School of MedicineIndianapolisINUSA
- Department of Pharmacology and ToxicologyIndiana University School of MedicineIndianapolisINUSA
- Indiana University Simon Comprehensive Cancer CenterIndiana University School of MedicineIndianapolisINUSA
| | - Melissa L. Fishel
- Department of Pediatrics and Herman B Wells Center for Pediatric ResearchIndiana University School of MedicineIndianapolisINUSA
- Department of Pharmacology and ToxicologyIndiana University School of MedicineIndianapolisINUSA
- Indiana University Simon Comprehensive Cancer CenterIndiana University School of MedicineIndianapolisINUSA
| |
Collapse
|
22
|
Torrens-Mas M, Roca P. Phytoestrogens for Cancer Prevention and Treatment. BIOLOGY 2020; 9:E427. [PMID: 33261116 PMCID: PMC7759898 DOI: 10.3390/biology9120427] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 11/21/2020] [Accepted: 11/25/2020] [Indexed: 12/17/2022]
Abstract
Phytoestrogens are a large group of natural compounds found in more than 300 plants. They have a close structural similarity to estrogens, which allow them to bind to both estrogen receptors (ER), ERα and ERβ, presenting a weak estrogenic activity. Phytoestrogens have been described as antioxidant, anti-inflammatory, anti-thrombotic, anti-allergic, and anti-tumoral agents. Their role in cancer prevention has been well documented, although their impact on treatment efficiency is controversial. Several reports suggest that phytoestrogens may interfere with the effect of anti-cancer drugs through the regulation of oxidative stress and other mechanisms. Furthermore, some phytoestrogens could exert a protective effect on healthy cells, thus reducing the secondary effects of cancer treatment. In this review, we have studied the recent research in this area to find evidence for the role of phytoestrogens in cancer prevention and therapy efficacy.
Collapse
Affiliation(s)
- Margalida Torrens-Mas
- Grupo Multidisciplinar de Oncología Traslacional, Institut Universitari d’Investigació en Ciències de la Salut, Universitat de les Illes Balears, 07122 Palma, Spain;
- Instituto de Investigación Sanitaria Illes Balears, 07010 Palma, Spain
| | - Pilar Roca
- Grupo Multidisciplinar de Oncología Traslacional, Institut Universitari d’Investigació en Ciències de la Salut, Universitat de les Illes Balears, 07122 Palma, Spain;
- Instituto de Investigación Sanitaria Illes Balears, 07010 Palma, Spain
- Ciber Fisiopatología Obesidad y Nutrición (CB06/03), Instituto Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
23
|
Rojo MÁ, Garrosa M, Jiménez P, Girbés T, Garcia-Recio V, Cordoba-Diaz M, Cordoba-Diaz D. Unexpected Toxicity of Green Tea Polyphenols in Combination with the Sambucus RIL Ebulin. Toxins (Basel) 2020; 12:E542. [PMID: 32842591 PMCID: PMC7551510 DOI: 10.3390/toxins12090542] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 08/12/2020] [Accepted: 08/20/2020] [Indexed: 11/17/2022] Open
Abstract
The safety of concentrated food complements intake is a major health concern. It has been well established that green tea polyphenols (GTPs) consumption promotes healthy effects. However, the ingestion of large amounts of GTPs is a matter of controversy due to reported adverse effects. We underwent a preliminary exploration of the effects of the oral administration of a standardized concentrated GTPs preparation on mice which suffered from reversible intestinal derangement promoted by sublethal amounts of the antiribosomal lectin ebulin f from dwarf elder (Sambucus ebulus L.). Neither independent oral administration of 30 mg/kg body weight Polyphenon 60 nor intraperitoneal administration of 2.5 mg/kg body weight ebulin f triggered lethal toxicity. In contrast, the simultaneous administration of these same doses of both Polyphenon 60 and ebulin f triggered an important and unexpected synergistic toxic action featured by the biphasic reduction of weight, which continued after eight days, reaching a reduction of 40%. Lethality appeared 2 days after the onset of the combined treatment and reached more than 50% after 10 days.
Collapse
Affiliation(s)
- M. Ángeles Rojo
- Area of Experimental Sciences, Miguel de Cervantes European University, 47012 Valladolid, Spain;
| | - Manuel Garrosa
- Area of Histology, Faculty of Medicine and INCYL, University of Valladolid, 47005 Valladolid, Spain;
| | - Pilar Jiménez
- Area of Nutrition and Food Sciences, Faculty of Medicine, University of Valladolid, 47005 Valladolid, Spain; (P.J.); (T.G.)
| | - Tomás Girbés
- Area of Nutrition and Food Sciences, Faculty of Medicine, University of Valladolid, 47005 Valladolid, Spain; (P.J.); (T.G.)
| | - Verónica Garcia-Recio
- Area of Pharmaceutics and Food Technology, Faculty of Pharmacy, Complutense University of Madrid, 28040 Madrid, Spain; (V.G.-R.); (M.C.-D.)
| | - Manuel Cordoba-Diaz
- Area of Pharmaceutics and Food Technology, Faculty of Pharmacy, Complutense University of Madrid, 28040 Madrid, Spain; (V.G.-R.); (M.C.-D.)
- University Institute of Industrial Pharmacy (IUFI), Complutense University of Madrid, 28040 Madrid, Spain
| | - Damián Cordoba-Diaz
- Area of Pharmaceutics and Food Technology, Faculty of Pharmacy, Complutense University of Madrid, 28040 Madrid, Spain; (V.G.-R.); (M.C.-D.)
- University Institute of Industrial Pharmacy (IUFI), Complutense University of Madrid, 28040 Madrid, Spain
| |
Collapse
|
24
|
Bose S, Banerjee S, Mondal A, Chakraborty U, Pumarol J, Croley CR, Bishayee A. Targeting the JAK/STAT Signaling Pathway Using Phytocompounds for Cancer Prevention and Therapy. Cells 2020; 9:E1451. [PMID: 32545187 PMCID: PMC7348822 DOI: 10.3390/cells9061451] [Citation(s) in RCA: 102] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 06/07/2020] [Accepted: 06/08/2020] [Indexed: 02/07/2023] Open
Abstract
Cancer is a prevalent cause of mortality around the world. Aberrated activation of Janus kinase (JAK)/signal transducer and activator of transcription (STAT) signaling pathway promotes tumorigenesis. Natural agents, including phytochemicals, exhibit potent anticancer activities via various mechanisms. However, the therapeutic potency of phytoconstituents as inhibitors of JAK/STAT signaling against cancer has only come into focus in recent days. The current review highlights phytochemicals that can suppress the JAK/STAT pathway in order to impede cancer cell growth. Various databases, such as PubMed, ScienceDirect, Web of Science, SpringerLink, Scopus, and Google Scholar, were searched using relevant keywords. Once the authors were in agreement regarding the suitability of a study, a full-length form of the relevant article was obtained, and the information was gathered and cited. All the complete articles that were incorporated after the literature collection rejection criteria were applied were perused in-depth and material was extracted based on the importance, relevance, and advancement of the apprehending of the JAK/STAT pathway and their relation to phytochemicals. Based on the critical and comprehensive analysis of literature presented in this review, phytochemicals from diverse plant origins exert therapeutic and cancer preventive effects, at least in part, through regulation of the JAK/STAT pathway. Nevertheless, more preclinical and clinical research is necessary to completely comprehend the capability of modulating JAK/STAT signaling to achieve efficient cancer control and treatment.
Collapse
Affiliation(s)
- Sankhadip Bose
- Department of Pharmacognosy, Bengal School of Technology, Chuchura 712 102, India;
| | - Sabyasachi Banerjee
- Department of Phytochemistry, Gupta College of Technological Sciences, Asansol 713 301, India; (S.B.); (U.C.)
| | - Arijit Mondal
- Department of Pharmaceutical Chemistry, Bengal College of Pharmaceutical Technology, Dubrajpur 731 123, India
| | - Utsab Chakraborty
- Department of Phytochemistry, Gupta College of Technological Sciences, Asansol 713 301, India; (S.B.); (U.C.)
| | - Joshua Pumarol
- Lake Erie College of Osteopathic Medicine, Bradenton, FL 34211, USA; (J.P.); (C.R.C.)
| | - Courtney R. Croley
- Lake Erie College of Osteopathic Medicine, Bradenton, FL 34211, USA; (J.P.); (C.R.C.)
| | - Anupam Bishayee
- Lake Erie College of Osteopathic Medicine, Bradenton, FL 34211, USA; (J.P.); (C.R.C.)
| |
Collapse
|
25
|
Polonio-Alcalá E, Palomeras S, Torres-Oteros D, Relat J, Planas M, Feliu L, Ciurana J, Ruiz-Martínez S, Puig T. Fatty Acid Synthase Inhibitor G28 Shows Anticancer Activity in EGFR Tyrosine Kinase Inhibitor Resistant Lung Adenocarcinoma Models. Cancers (Basel) 2020; 12:cancers12051283. [PMID: 32438613 PMCID: PMC7281741 DOI: 10.3390/cancers12051283] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 04/29/2020] [Accepted: 05/16/2020] [Indexed: 02/07/2023] Open
Abstract
Epidermal growth factor receptor (EGFR) tyrosine kinases inhibitors (TKIs) are effective therapies for non-small cell lung cancer (NSCLC) patients whose tumors harbor an EGFR activating mutation. However, this treatment is not curative due to primary and secondary resistance such as T790M mutation in exon 20. Recently, activation of transducer and activator of transcription 3 (STAT3) in NSCLC appeared as an alternative resistance mechanism allowing cancer cells to elude the EGFR signaling. Overexpression of fatty acid synthase (FASN), a multifunctional enzyme essential for endogenous lipogenesis, has been related to resistance and the regulation of the EGFR/Jak2/STAT signaling pathways. Using EGFR mutated (EGFRm) NSCLC sensitive and EGFR TKIs’ resistant models (Gefitinib Resistant, GR) we studied the role of the natural polyphenolic anti-FASN compound (−)-epigallocatechin-3-gallate (EGCG), and its derivative G28 to overcome EGFR TKIs’ resistance. We show that G28’s cytotoxicity is independent of TKIs’ resistance mechanisms displaying synergistic effects in combination with gefitinib and osimertinib in the resistant T790M negative (T790M−) model and showing a reduction of activated EGFR and STAT3 in T790M positive (T790M+) models. Our results provide the bases for further investigation of G28 in combination with TKIs to overcome the EGFR TKI resistance in NSCLC.
Collapse
Affiliation(s)
- Emma Polonio-Alcalá
- New Therapeutic Targets Laboratory (TargetsLab)-Oncology Unit, Department of Medical Sciences, Faculty of Medicine, University of Girona, 17003 Girona, Spain; (E.P.-A.); (S.P.)
- Product, Process and Production Engineering Research Group (GREP), Department of Mechanical Engineering and Industrial Construction, University of Girona, 17003 Girona, Spain;
| | - Sònia Palomeras
- New Therapeutic Targets Laboratory (TargetsLab)-Oncology Unit, Department of Medical Sciences, Faculty of Medicine, University of Girona, 17003 Girona, Spain; (E.P.-A.); (S.P.)
| | - Daniel Torres-Oteros
- Department of Nutrition, Food Sciences and Gastronomy, School of Pharmacy and Food Sciences, Food Torribera Campus, University of Barcelona, E-08921 Santa Coloma de Gramanet, Spain; (D.T.-O.); (J.R.)
| | - Joana Relat
- Department of Nutrition, Food Sciences and Gastronomy, School of Pharmacy and Food Sciences, Food Torribera Campus, University of Barcelona, E-08921 Santa Coloma de Gramanet, Spain; (D.T.-O.); (J.R.)
- Institute of Nutrition and Food Safety of the University of Barcelona (INSA-UB), E-08921 Santa Coloma de Gramenet, Spain
- CIBER Physiopathology of Obesity and Nutrition (CIBER-OBN), Instituto de Salud Carlos III, E-28029 Madrid, Spain
| | - Marta Planas
- LIPPSO, Department of Chemistry, University of Girona, 17003 Girona, Spain; (M.P.); (L.F.)
| | - Lidia Feliu
- LIPPSO, Department of Chemistry, University of Girona, 17003 Girona, Spain; (M.P.); (L.F.)
| | - Joaquim Ciurana
- Product, Process and Production Engineering Research Group (GREP), Department of Mechanical Engineering and Industrial Construction, University of Girona, 17003 Girona, Spain;
| | - Santiago Ruiz-Martínez
- New Therapeutic Targets Laboratory (TargetsLab)-Oncology Unit, Department of Medical Sciences, Faculty of Medicine, University of Girona, 17003 Girona, Spain; (E.P.-A.); (S.P.)
- Correspondence: (S.R.-M.); (T.P.); Tel.: +34-972-419-548 (S.R.-M.); +34-972-419-628 (T.P.)
| | - Teresa Puig
- New Therapeutic Targets Laboratory (TargetsLab)-Oncology Unit, Department of Medical Sciences, Faculty of Medicine, University of Girona, 17003 Girona, Spain; (E.P.-A.); (S.P.)
- Correspondence: (S.R.-M.); (T.P.); Tel.: +34-972-419-548 (S.R.-M.); +34-972-419-628 (T.P.)
| |
Collapse
|
26
|
Cao S, Han Y, Li Q, Chen Y, Zhu D, Su Z, Guo H. Mapping Pharmacological Network of Multi-Targeting Litchi Ingredients in Cancer Therapeutics. Front Pharmacol 2020. [DOI: 10.3389/fphar.2020.00451
expr 967555229 + 995954239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2023] Open
|
27
|
Cao S, Han Y, Li Q, Chen Y, Zhu D, Su Z, Guo H. Mapping Pharmacological Network of Multi-Targeting Litchi Ingredients in Cancer Therapeutics. Front Pharmacol 2020; 11:451. [PMID: 32390834 PMCID: PMC7193898 DOI: 10.3389/fphar.2020.00451] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Accepted: 03/23/2020] [Indexed: 12/12/2022] Open
Abstract
Considerable pharmacological studies have demonstrated that the extracts and ingredients from different parts (seeds, peels, pulps, and flowers) of Litchi exhibited anticancer effects by affecting the proliferation, apoptosis, autophagy, metastasis, chemotherapy and radiotherapy sensitivity, stemness, metabolism, angiogenesis, and immunity via multiple targeting. However, there is no systematical analysis on the interaction network of “multiple ingredients-multiple targets-multiple pathways” anticancer effects of Litchi. In this study, we summarized the confirmed anticancer ingredients and molecular targets of Litchi based on published articles and applied network pharmacology approach to explore the complex mechanisms underlying these effects from a perspective of system biology. The top ingredients, top targets, and top pathways of each anticancer function were identified using network pharmacology approach. Further intersecting analyses showed that Epigallocatechin gallate (EGCG), Gallic acid, Kaempferol, Luteolin, and Betulinic acid were the top ingredients which might be the key ingredients exerting anticancer function of Litchi, while BAX, BCL2, CASP3, and AKT1 were the top targets which might be the main targets underling the anticancer mechanisms of these top ingredients. These results provided references for further understanding and exploration of Litchi as therapeutics in cancer as well as the application of “Component Formula” based on Litchi’s effective ingredients.
Collapse
Affiliation(s)
- Sisi Cao
- College of Pharmacy, Guangxi Medical University, Nanning, China
| | - Yaoyao Han
- College of Pharmacy, Guangxi Medical University, Nanning, China.,Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education & Center for Translational Medicine, Guangxi Medical University, Nanning, China
| | - Qiaofeng Li
- Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education & Center for Translational Medicine, Guangxi Medical University, Nanning, China.,School of Preclinical Medicine, Guangxi Medical University, Nanning, China
| | - Yanjiang Chen
- Department of Surgery, University of Melbourne, Parkville, VIC, Australia
| | - Dan Zhu
- College of Pharmacy, Guangxi Medical University, Nanning, China
| | - Zhiheng Su
- College of Pharmacy, Guangxi Medical University, Nanning, China
| | - Hongwei Guo
- College of Pharmacy, Guangxi Medical University, Nanning, China.,Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education & Center for Translational Medicine, Guangxi Medical University, Nanning, China
| |
Collapse
|
28
|
Mohan CD, Rangappa S, Preetham HD, Chandra Nayaka S, Gupta VK, Basappa S, Sethi G, Rangappa KS. Targeting STAT3 signaling pathway in cancer by agents derived from Mother Nature. Semin Cancer Biol 2020; 80:157-182. [DOI: 10.1016/j.semcancer.2020.03.016] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 03/23/2020] [Accepted: 03/28/2020] [Indexed: 02/07/2023]
|
29
|
Induction of Endoplasmic Reticulum Stress Pathway by Green Tea Epigallocatechin-3-Gallate (EGCG) in Colorectal Cancer Cells: Activation of PERK/p-eIF2 α/ATF4 and IRE1 α. BIOMED RESEARCH INTERNATIONAL 2019; 2019:3480569. [PMID: 31930117 PMCID: PMC6942794 DOI: 10.1155/2019/3480569] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Accepted: 10/31/2019] [Indexed: 01/01/2023]
Abstract
Epigallocatechin-3-gallate (EGCG) is the most abundant bioactive polyphenolic compound among the green tea constituents and has been identified as a potential anticancer agent in colorectal cancer (CRC) studies. This study was aimed to determine the mechanism of actions of EGCG when targeting the endoplasmic reticulum (ER) stress pathway in CRC. The MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide) assay was performed on HT-29 cell line and normal cell line (3T3) to determine the EGCG toxicity. Next, western blot was done to observe the expression of the related proteins for the ER stress pathway. The Caspase 3/7 assay was performed to determine the apoptosis induced by EGCG. The results demonstrated that EGCG treatment was toxic to the HT-29 cell line. EGCG induced ER stress in HT-29 by upregulating immunoglobulin-binding (BiP), PKR-like endoplasmic reticulum kinase (PERK), phosphorylation of eukaryotic initiation factor 2 alpha subunit (eIF2α), activating transcription 4 (ATF4), and inositol-requiring kinase 1 alpha (IRE1α). Apoptosis was induced in HT-29 cells after the EGCG treatment, as shown by the Caspase 3/7 activity. This study indicates that green tea EGCG has the potential to inhibit colorectal cancer cells through the induction of ER stress.
Collapse
|
30
|
Wei R, Penso NEC, Hackman RM, Wang Y, Mackenzie GG. Epigallocatechin-3-Gallate (EGCG) Suppresses Pancreatic Cancer Cell Growth, Invasion, and Migration partly through the Inhibition of Akt Pathway and Epithelial-Mesenchymal Transition: Enhanced Efficacy when Combined with Gemcitabine. Nutrients 2019; 11:E1856. [PMID: 31405071 PMCID: PMC6722696 DOI: 10.3390/nu11081856] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 08/03/2019] [Accepted: 08/06/2019] [Indexed: 12/17/2022] Open
Abstract
Most pancreatic cancers are usually diagnosed at an advanced stage when they have already metastasized. Epigallocatechin-3-gallate (EGCG), a major polyphenolic constituent of green tea, has been shown to reduce pancreatic cancer growth, but its effect on metastasis remains elusive. This study evaluated the capacity of EGCG to inhibit pancreatic cancer cell migration and invasion and the underlying mechanisms. EGCG reduced pancreatic cancer cell growth, migration, and invasion in vitro and in vivo. EGCG prevented "Cadherin switch" and decreased the expression level of TCF8/ZEB1, β-Catenin, and Vimentin. Mechanistically, EGCG inhibited the Akt pathway in a time-dependent manner, by suppressing IGFR phosphorylation and inducing Akt degradation. Co-treatment with catalase or N-Acetyl-L-cysteine did not abrogate EGCG's effect on the Akt pathway or cell growth. Moreover, EGCG synergized with gemcitabine to suppress pancreatic cancer cell growth, migration, and invasion, through modulating epithelial-mesenchymal transition markers and inhibiting Akt pathway. In summary, EGCG may prove beneficial to improve gemcitabine sensitivity in inhibiting pancreatic cancer cell migration and invasion, to some extent through the inhibition of Akt pathway and epithelial-mesenchymal transition.
Collapse
Affiliation(s)
- Ran Wei
- Tea Science Institute, Zhejiang University, Hangzhou 310058, China
- Department of Nutrition, University of California, Davis, CA 95616, USA
| | | | - Robert M Hackman
- Department of Nutrition, University of California, Davis, CA 95616, USA
| | - Yuefei Wang
- Tea Science Institute, Zhejiang University, Hangzhou 310058, China.
| | | |
Collapse
|
31
|
Ma Y, Yu W, Shrivastava A, Srivastava RK, Shankar S. Inhibition of pancreatic cancer stem cell characteristics by α-Mangostin: Molecular mechanisms involving Sonic hedgehog and Nanog. J Cell Mol Med 2019; 23:2719-2730. [PMID: 30712329 PMCID: PMC6433724 DOI: 10.1111/jcmm.14178] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2018] [Revised: 12/31/2018] [Accepted: 01/02/2019] [Indexed: 12/21/2022] Open
Abstract
The current investigation was intended to elucidate the molecular mechanism of α‐Mangostin in the regulation of pancreatic cancer stem cell (CSC) characteristics. Here, we demonstrate that α‐Mangostin inhibited cell proliferation in pancreatic CSCs and cancer cell lines while it showed no effect on human pancreatic normal ductal epithelial cells. Also, α‐Mangostin inhibited colony formation and induced apoptosis in these cells. Further, α‐Mangostin inhibited the self‐renewal capacity of CSCs isolated from human primary tumours and KrasG12D mice. Furthermore, α‐Mangostin inhibited the invasive and metastatic ability of pancreatic CSCs by suppressing the epithelial‐to‐mesenchymal transition (EMT) via up‐regulation of E‐cadherin and down‐regulation of mesenchymal phenotype by inhibiting N‐cadherin, Snail and Slug expression. Interestingly, the pluripotency maintaining factors and CSC markers were inhibited by α‐Mangostin thus suggesting that α‐Mangostin can target CSCs to inhibit pancreatic cancer effectively. Gli signalling plays a crucial role in the self‐renewal and pluripotency of CSCs. α‐Mangostin inhibited the Gli transcription and the expression of Gli target genes (Nanog, Oct4, c‐Myc, Sox‐2 and KLF4) in CSCs. Using ChIP assay, we demonstrated that Nanog could directly bind to promoters of Cdk2, Cdk6, FGF4, c‐Myc and α‐Mangostin inhibited Nanog binding to these promoters. Conversely, the inhibitory effects of the α‐Mangostin on CSC proliferation and Gli or Nanog transcription and their targets were abrogated by either enforced activation of sonic hedgehog (Shh) or by the overexpression of Nanog. Taken together, our studies suggest that α‐Mangostin may act as Gli inhibitor and establishes the pre‐clinical significance of α‐Mangostin for the prevention and treatment of pancreatic cancer.
Collapse
Affiliation(s)
- Yiming Ma
- Kansas City VA Medical Center, Kansas City, Missouri
| | - Wei Yu
- Kansas City VA Medical Center, Kansas City, Missouri
| | - Anju Shrivastava
- Department of Oncology, St. Joseph's Hospital and Medical Center, Phoenix, Arizona
| | - Rakesh K Srivastava
- Kansas City VA Medical Center, Kansas City, Missouri.,Department of Pharmaceutical Sciences, University of Missouri-Kansas City, Kansas City, Missouri
| | - Sharmila Shankar
- Kansas City VA Medical Center, Kansas City, Missouri.,Department of Pathology, School of Medicine, University of Missouri-Kansas City, Kansas City, Missouri
| |
Collapse
|
32
|
Abotaleb M, Samuel SM, Varghese E, Varghese S, Kubatka P, Liskova A, Büsselberg D. Flavonoids in Cancer and Apoptosis. Cancers (Basel) 2018; 11:cancers11010028. [PMID: 30597838 PMCID: PMC6357032 DOI: 10.3390/cancers11010028] [Citation(s) in RCA: 371] [Impact Index Per Article: 61.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Revised: 12/12/2018] [Accepted: 12/13/2018] [Indexed: 12/19/2022] Open
Abstract
Cancer is the second leading cause of death globally. Although, there are many different approaches to cancer treatment, they are often painful due to adverse side effects and are sometimes ineffective due to increasing resistance to classical anti-cancer drugs or radiation therapy. Targeting delayed/inhibited apoptosis is a major approach in cancer treatment and a highly active area of research. Plant derived natural compounds are of major interest due to their high bioavailability, safety, minimal side effects and, most importantly, cost effectiveness. Flavonoids have gained importance as anti-cancer agents and have shown great potential as cytotoxic anti-cancer agents promoting apoptosis in cancer cells. In this review, a summary of flavonoids and their effectiveness in cancer treatment targeting apoptosis has been discussed.
Collapse
Affiliation(s)
- Mariam Abotaleb
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha, P.O. Box 24144, Qatar.
| | - Samson Mathews Samuel
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha, P.O. Box 24144, Qatar.
| | - Elizabeth Varghese
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha, P.O. Box 24144, Qatar.
| | - Sharon Varghese
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha, P.O. Box 24144, Qatar.
| | - Peter Kubatka
- Department of Medical Biology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 03601 Martin, Slovakia.
| | - Alena Liskova
- Clinic of Obstetrics and Gynecology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 03601 Martin, Slovakia.
| | - Dietrich Büsselberg
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha, P.O. Box 24144, Qatar.
| |
Collapse
|
33
|
Integration of phytochemicals and phytotherapy into cancer precision medicine. Oncotarget 2018; 8:50284-50304. [PMID: 28514737 PMCID: PMC5564849 DOI: 10.18632/oncotarget.17466] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Accepted: 02/18/2017] [Indexed: 01/01/2023] Open
Abstract
Concepts of individualized therapy in the 1970s and 1980s attempted to develop predictive in vitro tests for individual drug responsiveness without reaching clinical routine. Precision medicine attempts to device novel individual cancer therapy strategies. Using bioinformatics, relevant knowledge is extracted from huge data amounts. However, tumor heterogeneity challenges chemotherapy due to genetically and phenotypically different cell subpopulations, which may lead to refractory tumors. Natural products always served as vital resources for cancer therapy (e.g., Vinca alkaloids, camptothecin, paclitaxel, etc.) and are also sources for novel drugs. Targeted drugs developed to specifically address tumor-related proteins represent the basis of precision medicine. Natural products from plants represent excellent resource for targeted therapies. Phytochemicals and herbal mixtures act multi-specifically, i.e. they attack multiple targets at the same time. Network pharmacology facilitates the identification of the complexity of pharmacogenomic networks and new signaling networks that are distorted in tumors. In the present review, we give a conceptual overview, how the problem of drug resistance may be approached by integrating phytochemicals and phytotherapy into academic western medicine. Modern technology platforms (e.g. “-omics” technologies, DNA/RNA sequencing, and network pharmacology) can be applied for diverse treatment modalities such as cytotoxic and targeted chemotherapy as well as phytochemicals and phytotherapy. Thereby, these technologies represent an integrative momentum to merge the best of two worlds: clinical oncology and traditional medicine. In conclusion, the integration of phytochemicals and phytotherapy into cancer precision medicine represents a valuable asset to chemically synthesized chemicals and therapeutic antibodies.
Collapse
|
34
|
Green tea extract attenuates LPS-induced retinal inflammation in rats. Sci Rep 2018; 8:429. [PMID: 29323215 PMCID: PMC5765135 DOI: 10.1038/s41598-017-18888-5] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Accepted: 12/19/2017] [Indexed: 01/05/2023] Open
Abstract
Inflammation is in a wide spectrum of retinal diseases, causing irreversible blindness and visual impairment. We have previously demonstrated that Green Tea Extract (GTE) is a potent anti-inflammatory agent for anterior uveitis. Here we investigated the anti-inflammatory effect of GTE on lipopolysaccharides (LPS)-induced retinal inflammation in rats and explored the underlying mechanism. Adult rats were injected with LPS and GTE was administered intra-gastrically at 2, 8, 26 and 32 hours post-injection. Staining of whole-mount retina showed that the number of activated microglia cells was significantly increased at 48 hours post-injection, which was suppressed after GTE treatment in a dose-dependent manner. Activation of astrocytes and Müller glia in the retina was also suppressed after GTE treatment. Meanwhile, GTE reduced the expression of pro-inflammatory cytokines including IL-1β, TNF-α and IL-6 in retina and vitreous humor. These anti-inflammatory effects were associated with a reduced phosphorylation of STAT3 and NF-κB in the retina. Furthermore, the surface receptor of EGCG, 67LR, was localized on the neurons and glia in the retina. These findings demonstrate that GTE is an effective agent in suppressing LPS-induced retinal inflammation, probably through its potent anti-oxidative property and a receptor-mediated action on transcription factors that regulate production of pro-inflammatory cytokines.
Collapse
|
35
|
Docosahexaenoic acid inhibits 12-O-tetradecanoylphorbol-13- acetate-induced fascin-1-dependent breast cancer cell migration by suppressing the PKCδ- and Wnt-1/β-catenin-mediated pathways. Oncotarget 2018; 7:25162-79. [PMID: 27036017 PMCID: PMC5041895 DOI: 10.18632/oncotarget.7301] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Accepted: 01/27/2016] [Indexed: 11/25/2022] Open
Abstract
Fascin-1, an actin-bundling protein, plays an important role in cancer cell migration and invasion; however, the underlying mechanism remains unclear. On the basis of a 12-O-tetradecanoylphorbol 13-acetate (TPA)-induced cell migration model, it was shown that TPA increased fascin-1 mRNA and protein expression and fascin-1-dependent cell migration. TPA dose- and time-dependently increased PKCδ and STAT3α activation and GSK3β phosphorylation; up-regulated Wnt-1, β-catenin, and STAT3α expression; and increased the nuclear translocation of β-catenin and STAT3α. Rottlerin, a PKCδ inhibitor, abrogated the increases in STAT3α activation and β-catenin and fascin-1 expression. WP1066, a STAT3 inhibitor, suppressed TPA-induced STAT3α DNA binding activity and β-catenin expression. Knockdown of β-catenin attenuated TPA-induced fascin-1 and STAT3α expression as well as cell migration. In addition to MCF-7, migration of Hs578T breast cancer cells was inhibited by silencing fascin-1, β-catenin, and STAT3α expression as well. TPA also induced Wnt-1 expression and secretion, and blocking Wnt-1 signaling abrogated β-catenin induction. DHA pretreatment attenuated TPA-induced cell migration, PKCδ and STAT3α activation, GSK3β phosphorylation, and Wnt-1, β-catenin, STAT3α, and fascin-1 expression. Our results demonstrated that TPA-induced migration is likely associated with the PKCδ and Wnt-1 pathways, which lead to STAT3α activation, GSK3β inactivation, and β-catenin increase and up-regulation of fascin-1 expression. Moreover, the anti-metastatic potential of DHA is partly attributed to its suppression of TPA-activated PKCδ and Wnt-1 signaling.
Collapse
|
36
|
Rajagopal C, Lankadasari MB, Aranjani JM, Harikumar KB. Targeting oncogenic transcription factors by polyphenols: A novel approach for cancer therapy. Pharmacol Res 2018; 130:273-291. [PMID: 29305909 DOI: 10.1016/j.phrs.2017.12.034] [Citation(s) in RCA: 81] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Revised: 11/30/2017] [Accepted: 12/31/2017] [Indexed: 02/06/2023]
Abstract
Inflammation is one of the major causative factor of cancer and chronic inflammation is involved in all the major steps of cancer initiation, progression metastasis and drug resistance. The molecular mechanism of inflammation driven cancer is the complex interplay between oncogenic and tumor suppressive transcription factors which include FOXM1, NF-kB, STAT3, Wnt/β- Catenin, HIF-1α, NRF2, androgen and estrogen receptors. Several products derived from natural sources modulate the expression and activity of multiple transcription factors in various tumor models as evident from studies conducted in cell lines, pre-clinical models and clinical samples. Further combination of these natural products along with currently approved cancer therapies added an additional advantage and they considered as promising targets for prevention and treatment of inflammation and cancer. In this review we discuss the application of multi-targeting natural products by analyzing the literature and future directions for their plausible applications in drug discovery.
Collapse
Affiliation(s)
- Chitra Rajagopal
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram 695014, Kerala, India
| | - Manendra Babu Lankadasari
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram 695014, Kerala, India
| | - Jesil Mathew Aranjani
- Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| | - K B Harikumar
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram 695014, Kerala, India.
| |
Collapse
|
37
|
Combination curcumin and (-)-epigallocatechin-3-gallate inhibits colorectal carcinoma microenvironment-induced angiogenesis by JAK/STAT3/IL-8 pathway. Oncogenesis 2017; 6:e384. [PMID: 28967875 PMCID: PMC5668882 DOI: 10.1038/oncsis.2017.84] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Revised: 07/31/2017] [Accepted: 08/19/2017] [Indexed: 12/15/2022] Open
Abstract
Tumor microenvironment has a crucial role in cancer development and progression, whereas the mechanism of how it regulates angiogenesis is unclear. In this study, we simulated the colorectal carcinoma microenvironment by conditioned medium (CM) of colorectal carcinoma cell lines (SW620, HT-29, HCT116) supernatant or colorectal carcinoma tissue homogenate supernatant to induce normal endothelial cells (NECs). We found that colorectal carcinoma CM promoted tumor angiogenesis by coercing NECs toward tumor endothelial cells (TECs) with the activation of the JAK/STAT3 signaling pathway. Antibody array analysis showed HT-29 supernatant contained numerous angiogenesis-related proteins, especially IL-8. Interestingly, the production of IL-8 in NECs induced by HT-29 CM was also increased. We also verified the crucial role of IL-8 in promoting the CM-induced angiogenesis, as IL-8 repression by neutralizing antibody abolished the transition of NECs toward TECs. Curcumin and (-)-epigallocatechin-3-gallate (EGCG) are broadly investigated in cancer chemoprevention. However, poor bioavailability hurdles their application alone, and the mechanism of their anti-angiogenesis still need to be illuminated. Here, we found that curcumin combination with EGCG attenuated the tumor CM-induced transition of NECs toward TECs by inhibiting JAK/STAT3 signaling pathway. Furthermore, the combination of curcumin and EGCG markedly reduced tumor growth and angiogenesis in the colorectal carcinoma PDX mouse model, and the combined anti-angiogenic effect was better than that of curcumin or EGCG alone. Taken together, our findings provide a new mechanism of tumor angiogenesis, and the combination of curcumin and EGCG represents a potential anti-angiogenic therapeutic method for colorectal carcinoma.
Collapse
|
38
|
Bello M, Basilio-Antonio L, Fragoso-Vázquez J, Avalos-Soriano A, Correa-Basurto J. Molecular recognition between pancreatic lipase and natural and synthetic inhibitors. Int J Biol Macromol 2017; 98:855-868. [DOI: 10.1016/j.ijbiomac.2017.01.150] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Revised: 01/27/2017] [Accepted: 01/31/2017] [Indexed: 02/03/2023]
|
39
|
ErbB Proteins as Molecular Target of Dietary Phytochemicals in Malignant Diseases. JOURNAL OF ONCOLOGY 2017; 2017:1532534. [PMID: 28286519 PMCID: PMC5327764 DOI: 10.1155/2017/1532534] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Revised: 12/30/2016] [Accepted: 01/17/2017] [Indexed: 12/16/2022]
Abstract
ErbB proteins overexpression, in both normal and mutated forms, is associated with invasive forms of cancer prone to metastasis and with stronger antiapoptotic mechanisms and therefore more challenging to treat. Downstream effectors of ErbB receptors mediating these phenotypic traits include MAPK, STAT, and PI3K/AKT/mTOR pathways. Various phytochemical compounds were studied for their large number of biological effects including anticancer activity. Among these compounds, epigallocatechin-3-gallate (EGCG), the main catechin from green tea leaves, and curcumin, component of the curry powder, constituted the object of numerous studies. Both compounds were shown to act directly either on ErbB expression, or on their downstream signaling molecules. In this paper we aim to review the involvement of ErbB proteins in cancer as well as the biologic activity of EGCG and curcumin in ErbB expressing and overexpressing malignancies. The problems arising in the administration of the two compounds due to their reduced bioavailability when orally administered, as well as the progress made in this field, from using novel formulations to improved dosing regimens or improved synthetic analogs, are also discussed.
Collapse
|
40
|
Cancer prevention and therapy through the modulation of transcription factors by bioactive natural compounds. Semin Cancer Biol 2016; 40-41:35-47. [DOI: 10.1016/j.semcancer.2016.03.005] [Citation(s) in RCA: 162] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Revised: 03/29/2016] [Accepted: 03/30/2016] [Indexed: 02/07/2023]
|
41
|
Eriksson E, Wenthe J, Irenaeus S, Loskog A, Ullenhag G. Gemcitabine reduces MDSCs, tregs and TGFβ-1 while restoring the teff/treg ratio in patients with pancreatic cancer. J Transl Med 2016; 14:282. [PMID: 27687804 PMCID: PMC5041438 DOI: 10.1186/s12967-016-1037-z] [Citation(s) in RCA: 141] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Accepted: 09/21/2016] [Indexed: 02/06/2023] Open
Abstract
Background Cancer immunotherapy can be potentiated by conditioning regimens such as cyclophosphamide, which reduces the level of regulatory T cells (tregs). However, myeloid suppressive cells are still remaining. Accordingly to previous reports, gemcitabine improves immune status of cancer patients. In this study, the role of gemcitabine was further explored to map its immunological target cells and molecules in patients with pancreatic cancer. Methods Patient blood was investigated by flow cytometry and cytokine arrays at different time points during gemcitabine treatment. Results The patients had elevated myeloid-derived suppressor cells (MDSCs), and Tregs at diagnosis. Myeloid cells were in general decreased by gemcitabine. The granulocytic MDSCs were significantly reduced while monocytic MDSCs were not affected. In vitro, monocytes responding to IL-6 by STAT3 phosphorylation were prevented to respond in gemcitabine medium. However, gemcitabine could not prevent STAT3 phosphorylation in IL-6-treated tumor cell lines. TGFβ-1 was significantly reduced after only one treatment and continued to decrease. At the same time, the effector T cell:Treg ratio was increased and the effector T cells had full proliferative capacity during the gemcitabine cycle. However, after a resting period, the level of suppressor cells and TGFβ-1 had been restored showing the importance of continuous conditioning. Conclusions Gemcitabine regulates the immune system in patients with pancreatic cancer including MDSCs, Tregs and molecules such as TGFβ-1 but does not hamper the ability of effector lymphocytes to expand to stimuli. Hence, it may be of high interest to use gemcitabine as a conditioning strategy together with immunotherapy. Electronic supplementary material The online version of this article (doi:10.1186/s12967-016-1037-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Emma Eriksson
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Rudbeck Laboratory C11 2nd floor, Uppsala University, Dag Hammarskjoldsvag 20, 751 85, Uppsala, Sweden
| | - Jessica Wenthe
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Rudbeck Laboratory C11 2nd floor, Uppsala University, Dag Hammarskjoldsvag 20, 751 85, Uppsala, Sweden
| | - Sandra Irenaeus
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Rudbeck Laboratory C11 2nd floor, Uppsala University, Dag Hammarskjoldsvag 20, 751 85, Uppsala, Sweden.,Department of Oncology, Uppsala University Hospital, Uppsala, Sweden
| | - Angelica Loskog
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Rudbeck Laboratory C11 2nd floor, Uppsala University, Dag Hammarskjoldsvag 20, 751 85, Uppsala, Sweden. .,Lokon Pharma AB, Uppsala, Sweden.
| | - Gustav Ullenhag
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Rudbeck Laboratory C11 2nd floor, Uppsala University, Dag Hammarskjoldsvag 20, 751 85, Uppsala, Sweden.,Department of Oncology, Uppsala University Hospital, Uppsala, Sweden
| |
Collapse
|
42
|
Sharma V, Kumar L, Mohanty SK, Maikhuri JP, Rajender S, Gupta G. Sensitization of androgen refractory prostate cancer cells to anti-androgens through re-expression of epigenetically repressed androgen receptor - Synergistic action of quercetin and curcumin. Mol Cell Endocrinol 2016; 431:12-23. [PMID: 27132804 DOI: 10.1016/j.mce.2016.04.024] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Revised: 04/02/2016] [Accepted: 04/27/2016] [Indexed: 01/06/2023]
Abstract
Epigenetic repression of Androgen Receptor (AR) gene by hypermethylation of its promoter causes resistance in prostate cancer (CaP) to androgen deprivation therapy with anti-androgens. Some dietary phytocompounds like quercetin (Q) and curcumin (C) with reported DNMT-inhibitory activity were tested for their ability to re-express the AR in AR-negative CaP cell lines PC3 and DU145. Combined treatment with Q+C was much more effective than either Q or C in inhibiting DNMT, causing global hypomethylation, restoring AR mRNA and protein levels and causing apoptosis via mitochondrial depolarization of PC3 and DU145. The functional AR protein expressed in Q+C treated cells sensitized them to dihydrotestosterone (DHT)-induced proliferation, bicalutamide-induced apoptosis, bound to androgen response element to increase luciferase activity in gene reporter assay and was susceptible to downregulation by AR siRNA. Bisulfite sequencing revealed high methylation of AR promoter CpG sites in AR-negative DU145 and PC3 cell lines that was significantly demethylated by Q+C treatment, which restored AR expression. Notable synergistic effects of Q+C combination in re-sensitizing androgen refractory CaP cells to AR-mediated apoptosis, their known safety in clinical use, and epidemiological evidences relating their dietary consumption with lower cancer incidences indicate their potential for use in chemoprevention of androgen resistance in prostate cancer.
Collapse
Affiliation(s)
- Vikas Sharma
- Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Lokesh Kumar
- Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Sujit K Mohanty
- Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Jagdamba P Maikhuri
- Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Singh Rajender
- Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Gopal Gupta
- Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow 226031, India.
| |
Collapse
|
43
|
Lee SY, Jung YO, Ryu JG, Oh HJ, Son HJ, Lee SH, Kwon JE, Kim EK, Park MK, Park SH, Kim HY, Cho ML. Epigallocatechin-3-gallate ameliorates autoimmune arthritis by reciprocal regulation of T helper-17 regulatory T cells and inhibition of osteoclastogenesis by inhibiting STAT3 signaling. J Leukoc Biol 2016; 100:559-68. [PMID: 26957211 DOI: 10.1189/jlb.3a0514-261rr] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2014] [Accepted: 02/08/2016] [Indexed: 11/24/2022] Open
Abstract
The green tea polyphenol epigallocatechin-3-gallate is a potent antioxidant. Here, we describe the effects of epigallocatechin-3-gallate on T cell differentiation and osteoclast differentiation in an animal model of arthritis. Mice with collagen-induced arthritis were injected intraperitoneally with epigallocatechin-3-gallate, 3 times/wk after the primary immunization. Surface markers of T helper 17 cells and regulatory T cells were analyzed by flow cytometry. Flow cytometry, Western blotting, and enzyme-linked immunosorbent assays were used to evaluate the effect of epigallocatechin-3-gallate on cell signaling in the collagen-induced arthritis model. Epigallocatechin-3-gallate decreased the arthritis index and showed protective effects against joint destruction in collagen-induced arthritis mice. The expression of cytokines, oxidative stress proteins, and phosphorylated-signal transducer and activator of transcription-3, 705 and 727, were significantly less in mice treated with epigallocatechin-3-gallate than it was in controls. Epigallocatechin-3-gallate reduced the expression of osteoclast markers in vitro and in vivo relative to the control, and the antiosteoclastic activity was observed in epigallocatechin-3-gallate-treated, interferon-γ knockout mice. The proportion of forkhead box protein 3-positive regulatory T cells was increased in the spleens of mice treated with epigallocatechin-3-gallate compared with control mice, whereas the proportion of T helper 17 cells was reduced. In vitro, the expression of nuclear respiratory factor 2, heme oxygenase-1, and extracellular signal-regulated kinase was increased significantly by epigallocatechin-3-gallate. We demonstrated that the administration of epigallocatechin-3-gallate attenuated the symptoms of arthritis, inhibited osteoclastogenesis and T helper 17 cell activation, and increased the number of regulatory T cells. At the molecular level, the antiarthritic effects of epigallocatechin-3-gallate may be due to induction of phosphorylated-extracellular signal-regulated kinase, nuclear respiratory factor 2, and heme oxygenase-1 and inhibition of signal transducer and activator of transcription-3 activation.
Collapse
Affiliation(s)
- Seon-Yeong Lee
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, The Catholic University of Korea, Banpo-dong, Seocho-gu, Seoul, South Korea
| | - Young Ok Jung
- Division of Rheumatology, Department of Internal Medicine, Hallym University Kang-Nam Sacred Heart Hospital, Seoul, Korea
| | - Jun-Geol Ryu
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, The Catholic University of Korea, Banpo-dong, Seocho-gu, Seoul, South Korea
| | - Hye-Jwa Oh
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, The Catholic University of Korea, Banpo-dong, Seocho-gu, Seoul, South Korea
| | - Hye-Jin Son
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, The Catholic University of Korea, Banpo-dong, Seocho-gu, Seoul, South Korea
| | - Seung Hoon Lee
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, The Catholic University of Korea, Banpo-dong, Seocho-gu, Seoul, South Korea
| | - Jeong-Eun Kwon
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, The Catholic University of Korea, Banpo-dong, Seocho-gu, Seoul, South Korea
| | - Eun-Kyung Kim
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, The Catholic University of Korea, Banpo-dong, Seocho-gu, Seoul, South Korea
| | - Mi-Kyung Park
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, The Catholic University of Korea, Banpo-dong, Seocho-gu, Seoul, South Korea
| | - Sung-Hwan Park
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, The Catholic University of Korea, Banpo-dong, Seocho-gu, Seoul, South Korea; Center for Rheumatic Disease, Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea, Banpo-dong, Seocho-gu, Seoul, South Korea; and
| | - Ho-Youn Kim
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, The Catholic University of Korea, Banpo-dong, Seocho-gu, Seoul, South Korea; Center for Rheumatic Disease, Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea, Banpo-dong, Seocho-gu, Seoul, South Korea; and
| | - Mi-La Cho
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, The Catholic University of Korea, Banpo-dong, Seocho-gu, Seoul, South Korea;
| |
Collapse
|
44
|
Blockage of STAT3 Signaling Pathway by Morusin Induces Apoptosis and Inhibits Invasion in Human Pancreatic Tumor Cells. Pancreas 2016; 45:409-19. [PMID: 26646273 DOI: 10.1097/mpa.0000000000000496] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
OBJECTIVES Signal transducer and activator of transcription 3 (STAT3) is an oncogenic transcription factor implicated in carcinogenesis. Here, we investigated the role of morusin, the major prenylflavonoid, isolated from Chinese herbal medicine in abrogating the constitutive STAT3 activation in human pancreatic tumor cells. METHODS The effect of morusin on STAT3 activation, associated protein kinases, STAT3-regulated gene products, cellular proliferation, and apoptosis was examined. RESULTS Morusin specifically inhibited constitutive STAT3 activation both at tyrosine residue 705 and serine residue 727 in 4 pancreatic tumor cells. The inhibition of STAT3 was mediated through the suppression of activation of upstream JAK1, JAK2, and c-Src kinases. Morusin led to the accumulation of the cells in different phases of the cell cycle and caused induction of apoptosis and loss of mitochondrial membrane potential. Morusin downregulated the expression of various STAT3-regulated gene products; this correlated with induction of caspase-3 activation and anti-invasive effects. Treatment with the protein tyrosine phosphatase inhibitor pervanadate reversed the morusin-induced downregulation of STAT3, thereby suggesting the involvement of a protein tyrosine phosphatase. CONCLUSIONS Morusin is a novel blocker of STAT3 activation and thus may have potential in negative regulation of growth and metastasis of pancreatic tumor cells.
Collapse
|
45
|
The Role of miRNAs in the Regulation of Pancreatic Cancer Stem Cells. Stem Cells Int 2016; 2016:8352684. [PMID: 27006664 PMCID: PMC4783541 DOI: 10.1155/2016/8352684] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2015] [Revised: 12/24/2015] [Accepted: 01/27/2016] [Indexed: 12/17/2022] Open
Abstract
Pancreatic ductal adenocarcinoma is currently one of the deadliest cancers with low overall survival rate. This disease leads to an aggressive local invasion and early metastases and is poorly responsive to treatment with chemotherapy or chemoradiotherapy. Several studies have shown that pancreatic cancer stem cells (PCSCs) play different roles in the regulation of drug resistance and recurrence in pancreatic cancer. MicroRNA (miRNA), a class of newly emerging small noncoding RNAs, is involved in the modulation of several biological activities ranging from invasion to metastases development, as well as drug resistance of pancreatic cancer. In this review, we synthesize the latest findings on the role of miRNAs in regulating different biological properties of pancreatic cancer stem cells.
Collapse
|
46
|
Oliveira MRD, Nabavi SF, Daglia M, Rastrelli L, Nabavi SM. Epigallocatechin gallate and mitochondria-A story of life and death. Pharmacol Res 2015; 104:70-85. [PMID: 26731017 DOI: 10.1016/j.phrs.2015.12.027] [Citation(s) in RCA: 111] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Revised: 12/20/2015] [Accepted: 12/23/2015] [Indexed: 01/05/2023]
Abstract
Epigallocatechin gallate (EGCG) is a flavonoid belonging to the chemical class of falvan-3-ols (catechins) esterified with gallic acid. It is the main catechin found in green tea (Camellia sinensis L.) accounting for about 50% of its total polyphenols. Extensive research performed in recent years has revealed that green tea demonstrates a wide range of positive biological activities against serious chronic diseases such as cardiovascular and neurodegenerative pathologies, cancer, metabolic syndrome and type 2 diabetes. These protective properties can be traced back to the potent antioxidant and anti-inflammatory activities of EGCG. Recent studies have suggested that it may exert its beneficial effects by modulating mitochondrial functions impacting mitochondrial biogenesis, bioenergetic control (ATP production and anabolism), alteration of the cell cycle, and mitochondria-related apoptosis. This review evaluates recent evidence on the ability of EGCG to exert critical influence on the above mentioned pathways.
Collapse
Affiliation(s)
- Marcos Roberto de Oliveira
- Department of Chemistry, ICET, Federal University of Mato Grosso (UFMT), Av. Fernando Corrêa da Costa, 2367, CEP 78060-900 Cuiabá, MT, Brazil.
| | - Seyed Fazel Nabavi
- Applied Biotechnology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Maria Daglia
- Department of Drug Sciences, Medicinal Chemistry and Pharmaceutical Technology Section, University of Pavia, Italy
| | - Luca Rastrelli
- Dipartimento di Farmacia, University of Salerno, via Giovanni Paolo II, 84084 Fisciano, Italy
| | - Seyed Mohammad Nabavi
- Applied Biotechnology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| |
Collapse
|
47
|
Zhang P, Yang B, Yao YY, Zhong LX, Chen XY, Kong QY, Wu ML, Li C, Li H, Liu J. PIAS3, SHP2 and SOCS3 Expression patterns in Cervical Cancers: Relevance with activation and resveratrol-caused inactivation of STAT3 signaling. Gynecol Oncol 2015; 139:529-35. [DOI: 10.1016/j.ygyno.2015.09.087] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Revised: 09/21/2015] [Accepted: 09/28/2015] [Indexed: 01/19/2023]
|
48
|
Casey SC, Amedei A, Aquilano K, Azmi AS, Benencia F, Bhakta D, Bilsland AE, Boosani CS, Chen S, Ciriolo MR, Crawford S, Fujii H, Georgakilas AG, Guha G, Halicka D, Helferich WG, Heneberg P, Honoki K, Keith WN, Kerkar SP, Mohammed SI, Niccolai E, Nowsheen S, Vasantha Rupasinghe HP, Samadi A, Singh N, Talib WH, Venkateswaran V, Whelan RL, Yang X, Felsher DW. Cancer prevention and therapy through the modulation of the tumor microenvironment. Semin Cancer Biol 2015; 35 Suppl:S199-S223. [PMID: 25865775 PMCID: PMC4930000 DOI: 10.1016/j.semcancer.2015.02.007] [Citation(s) in RCA: 251] [Impact Index Per Article: 27.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2014] [Revised: 02/26/2015] [Accepted: 02/27/2015] [Indexed: 02/06/2023]
Abstract
Cancer arises in the context of an in vivo tumor microenvironment. This microenvironment is both a cause and consequence of tumorigenesis. Tumor and host cells co-evolve dynamically through indirect and direct cellular interactions, eliciting multiscale effects on many biological programs, including cellular proliferation, growth, and metabolism, as well as angiogenesis and hypoxia and innate and adaptive immunity. Here we highlight specific biological processes that could be exploited as targets for the prevention and therapy of cancer. Specifically, we describe how inhibition of targets such as cholesterol synthesis and metabolites, reactive oxygen species and hypoxia, macrophage activation and conversion, indoleamine 2,3-dioxygenase regulation of dendritic cells, vascular endothelial growth factor regulation of angiogenesis, fibrosis inhibition, endoglin, and Janus kinase signaling emerge as examples of important potential nexuses in the regulation of tumorigenesis and the tumor microenvironment that can be targeted. We have also identified therapeutic agents as approaches, in particular natural products such as berberine, resveratrol, onionin A, epigallocatechin gallate, genistein, curcumin, naringenin, desoxyrhapontigenin, piperine, and zerumbone, that may warrant further investigation to target the tumor microenvironment for the treatment and/or prevention of cancer.
Collapse
Affiliation(s)
- Stephanie C Casey
- Division of Oncology, Departments of Medicine and Pathology, Stanford University School of Medicine, Stanford, CA, United States
| | - Amedeo Amedei
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Katia Aquilano
- Department of Biology, University of Rome "Tor Vergata", Rome, Italy
| | - Asfar S Azmi
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI, United States
| | - Fabian Benencia
- Department of Biomedical Sciences, Ohio University, Athens, OH, United States
| | - Dipita Bhakta
- School of Chemical and Biotechnology, SASTRA University, Thanjavur 613401, Tamil Nadu, India
| | - Alan E Bilsland
- Institute of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Chandra S Boosani
- Department of Biomedical Sciences, School of Medicine, Creighton University, Omaha, NE, United States
| | - Sophie Chen
- Ovarian and Prostate Cancer Research Laboratory, Guildford, Surrey, United Kingdom
| | | | - Sarah Crawford
- Department of Biology, Southern Connecticut State University, New Haven, CT, United States
| | - Hiromasa Fujii
- Department of Orthopedic Surgery, Nara Medical University, Kashihara, Japan
| | - Alexandros G Georgakilas
- Physics Department, School of Applied Mathematics and Physical Sciences, National Technical University of Athens, Athens, Greece
| | - Gunjan Guha
- School of Chemical and Biotechnology, SASTRA University, Thanjavur 613401, Tamil Nadu, India
| | | | - William G Helferich
- University of Illinois at Urbana-Champaign, Champaign-Urbana, IL, United States
| | - Petr Heneberg
- Charles University in Prague, Third Faculty of Medicine, Prague, Czech Republic
| | - Kanya Honoki
- Department of Orthopedic Surgery, Nara Medical University, Kashihara, Japan
| | - W Nicol Keith
- Institute of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Sid P Kerkar
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Sulma I Mohammed
- Department of Comparative Pathobiology, Purdue University Center for Cancer Research, West Lafayette, IN, United States
| | | | - Somaira Nowsheen
- Medical Scientist Training Program, Mayo Graduate School, Mayo Medical School, Mayo Clinic, Rochester, MN, United States
| | - H P Vasantha Rupasinghe
- Department of Environmental Sciences, Faculty of Agriculture, Dalhousie University, Nova Scotia, Canada
| | | | - Neetu Singh
- Advanced Molecular Science Research Centre (Centre for Advanced Research), King George's Medical University, Lucknow, Uttar Pradesh, India
| | - Wamidh H Talib
- Department of Clinical Pharmacy and Therapeutics, Applied Science University, Amman, Jordan
| | | | - Richard L Whelan
- Mount Sinai Roosevelt Hospital, Icahn Mount Sinai School of Medicine, New York City, NY, United States
| | - Xujuan Yang
- University of Illinois at Urbana-Champaign, Champaign-Urbana, IL, United States
| | - Dean W Felsher
- Division of Oncology, Departments of Medicine and Pathology, Stanford University School of Medicine, Stanford, CA, United States.
| |
Collapse
|
49
|
Singh BN, Singh HB, Singh A, Naqvi AH, Singh BR. Dietary phytochemicals alter epigenetic events and signaling pathways for inhibition of metastasis cascade: phytoblockers of metastasis cascade. Cancer Metastasis Rev 2015; 33:41-85. [PMID: 24390421 DOI: 10.1007/s10555-013-9457-1] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Cancer metastasis is a multistep process in which a cancer cell spreads from the site of the primary lesion, passes through the circulatory system, and establishes a secondary tumor at a new nonadjacent organ or part. Inhibition of cancer progression by dietary phytochemicals (DPs) offers significant promise for reducing the incidence and mortality of cancer. Consumption of DPs in the diet has been linked to a decrease in the rate of metastatic cancer in a number of preclinical animal models and human epidemiological studies. DPs have been reported to modulate the numerous biological events including epigenetic events (noncoding micro-RNAs, histone modification, and DNA methylation) and multiple signaling transduction pathways (Wnt/β-catenin, Notch, Sonic hedgehog, COX-2, EGFR, MAPK-ERK, JAK-STAT, Akt/PI3K/mTOR, NF-κB, AP-1, etc.), which can play a key role in regulation of metastasis cascade. Extensive studies have also been performed to determine the molecular mechanisms underlying antimetastatic activity of DPs, with results indicating that these DPs have significant inhibitory activity at nearly every step of the metastatic cascade. DPs have anticancer effects by inducing apoptosis and by inhibiting cell growth, migration, invasion, and angiogenesis. Growing evidence has also shown that these natural agents potentiate the efficacy of chemotherapy and radiotherapy through the regulation of multiple signaling pathways. In this review, we discuss the variety of molecular mechanisms by which DPs regulate metastatic cascade and highlight the potentials of these DPs as promising therapeutic inhibitors of cancer.
Collapse
Affiliation(s)
- B N Singh
- Research and Development Division, Sowbhagya Biotech Private Limited, Cherlapally, Hyderabad, 500051, Andhra Pradesh, India
| | | | | | | | | |
Collapse
|
50
|
Jung JH, Yun M, Choo EJ, Kim SH, Jeong MS, Jung DB, Lee H, Kim EO, Kato N, Kim B, Srivastava SK, Kaihatsu K, Kim SH. A derivative of epigallocatechin-3-gallate induces apoptosis via SHP-1-mediated suppression of BCR-ABL and STAT3 signalling in chronic myelogenous leukaemia. Br J Pharmacol 2015; 172:3565-78. [PMID: 25825203 DOI: 10.1111/bph.13146] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2014] [Revised: 03/15/2015] [Accepted: 03/23/2015] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND AND PURPOSE Epigallocatechin-3-gallate (EGCG) is a component of green tea known to have chemo-preventative effects on several cancers. However, EGCG has limited clinical application, which necessitates the development of a more effective EGCG prodrug as an anticancer agent. EXPERIMENTAL APPROACH Derivatives of EGCG were evaluated for their stability and anti-tumour activity in human chronic myeloid leukaemia (CML) K562 and KBM5 cells. KEY RESULTS EGCG-mono-palmitate (EGCG-MP) showed most prolonged stability compared with other EGCG derivatives. EGCG-MP exerted greater cytotoxicity and apoptosis in K562 and KBM5 cells than the other EGCG derivatives. EGCG-MP induced Src-homology 2 domain-containing tyrosine phosphatase 1 (SHP-1) leading decreased oncogenic protein BCR-ABL and STAT3 phosphorylation in CML cells, compared with treatment with EGCG. Furthermore, EGCG-MP reduced phosphorylation of STAT3 and survival genes in K562 cells, compared with EGCG. Conversely, depletion of SHP-1 or application of the tyrosine phosphatase inhibitor pervanadate blocked the ability of EGCG-MP to suppress phosphorylation of BCR-ABL and STAT3, and the expression of survival genes downstream of STAT3. In addition, EGCG-MP treatment more effectively suppressed tumour growth in BALB/c athymic nude mice compared with untreated controls or EGCG treatment. Immunohistochemistry revealed increased caspase 3 and SHP-1 activity and decreased phosphorylation of BCR-ABL in the EGCG-MP-treated group relative to that in the EGCG-treated group. CONCLUSIONS AND IMPLICATIONS EGCG-MP induced SHP-1-mediated inhibition of BCR-ABL and STAT3 signalling in vitro and in vivo more effectively than EGCG. This derivative may be a potent chemotherapeutic agent for CML treatment.
Collapse
Affiliation(s)
- Ji Hoon Jung
- College of Korean Medicine, Kyung Hee University, Seoul, South Korea
| | - Miyong Yun
- College of Korean Medicine, Kyung Hee University, Seoul, South Korea
| | - Eun-Jeong Choo
- College of Korean Medicine, Kyung Hee University, Seoul, South Korea
| | - Sun-Hee Kim
- College of Korean Medicine, Kyung Hee University, Seoul, South Korea
| | - Myoung-Seok Jeong
- College of Korean Medicine, Kyung Hee University, Seoul, South Korea
| | - Deok-Beom Jung
- College of Korean Medicine, Kyung Hee University, Seoul, South Korea
| | - Hyemin Lee
- College of Korean Medicine, Kyung Hee University, Seoul, South Korea
| | - Eun-Ok Kim
- College of Korean Medicine, Kyung Hee University, Seoul, South Korea
| | - Nobuo Kato
- The Institute of Scientific and Industrial Research, Osaka University, Osaka, Japan
| | - Bonglee Kim
- College of Korean Medicine, Kyung Hee University, Seoul, South Korea
| | - Sanjay K Srivastava
- Department of Biomedical Sciences and Cancer Biology Center, Texas Tech University Health Sciences Center, Amarillo, TX, USA
| | - Kunihiro Kaihatsu
- The Institute of Scientific and Industrial Research, Osaka University, Osaka, Japan
| | - Sung-Hoon Kim
- College of Korean Medicine, Kyung Hee University, Seoul, South Korea
| |
Collapse
|