1
|
Chen XF, Liu PG, Sheng N, Li XS, Hu RK, Zhu LX, Feng P. Arctigenin inhibits the progression of colorectal cancer through epithelial-mesenchymal transition via PI3K/Akt/mTOR signaling pathway. PLoS One 2024; 19:e0308947. [PMID: 39331595 PMCID: PMC11432899 DOI: 10.1371/journal.pone.0308947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 08/01/2024] [Indexed: 09/29/2024] Open
Abstract
BACKGROUND Colorectal cancer (CRC) is a significant disease worldwide, with high mortality rates. Conventional treatment methods often lead to metastasis and drug resistance, highlighting the need to explore new drugs and their potential molecular mechanisms. In this study, we investigated the effects of arctigenin on CRC cell proliferation, migration, invasion, apoptosis, and related protein expression, as well as its potential molecular mechanisms. METHODS The CCK-8 assay, transwell migration and invasion assays, flow cytometry, immunoblotting and immunofluorescence staining, western blot and an allograft tumor transplantation model was used. RESULTS Our study revealed that arctigenin effectively inhibited CRC cell proliferation, migration, and invasion in a dose-dependent manner, while also inducing apoptosis. At the molecular level, arctigenin significantly downregulated the expressions of PCNA, Bcl2, MMP-2, and MMP-9 and upregulated the expressions of Bax and cleaved caspase-3. Additionally, arctigenin demonstrated the ability to inhibit the epithelial-mesenchymal transition (EMT) process by upregulating E-cadherin and downregulating mesenchymal markers, such as N-cadherin, Vimentin, Snail, and Slug. Furthermore, arctigenin could inhibit the activation of the PI3K-AKT-mTOR signaling pathway, which has been implicated in cancer progression. In vivo experiments also showed that arctigenin significantly reduced tumor volume and size compared to the control group, with no significant adverse effects on the liver. CONCLUSIONS This is the first study to elucidate the mechanism by which arctigenin inhibits colorectal cancer metastasis through the PI3K-AKT-mTOR signaling pathway by suppressing the EMT process at the molecular level.
Collapse
Affiliation(s)
- Xiang-Fan Chen
- Biological Sample Bank, Afliated Hospital 2 of Nantong University, Nantong, China
| | - Pei-Gen Liu
- Department of General Surgery, Central Hospital of Panzhihua City, Panzhihua, Sichuan Province, China
| | - Nan Sheng
- Department of Clinical Laboratory, Afliated Hospital 2 of Nantong University, Nantong, China
| | - Xin-Shuai Li
- Department of Pharmacy, Afliated Hospital 2 of Nantong University, Nantong, China
| | - Rui-Kun Hu
- Personnel Department, Affiliated Maternity and Child Health Care Hospital of Nantong University, Nantong, China
| | - Long-Xun Zhu
- Department of Pharmacy, Afliated Hospital 2 of Nantong University, Nantong, China
| | - Panfeng Feng
- Department of Pharmacy, Afliated Hospital 2 of Nantong University, Nantong, China
| |
Collapse
|
2
|
Zhang P, Zhou C, Jing Q, Gao Y, Yang L, Li Y, Du J, Tong X, Wang Y. Role of APR3 in cancer: apoptosis, autophagy, oxidative stress, and cancer therapy. Apoptosis 2023; 28:1520-1533. [PMID: 37634193 DOI: 10.1007/s10495-023-01882-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/05/2023] [Indexed: 08/29/2023]
Abstract
APR3 (Apoptosis-related protein 3) is a gene that has recently been identified to be associated with apoptosis. The gene is located on human chromosome 2p22.3 and contains both transmembrane and EGF (epidermal growth factor)-like domains. Additionally, it has structural sites, including AP1, SP1, and MEF2D, that indicate NFAT (nuclear factor of activated T cells) and NF-κB (nuclear factor kappa-B) may be transcription factors for this gene. Functionally, APR3 participates in apoptosis due to the induction of mitochondrial damage to release mitochondrial cytochrome C. Concurrently, APR3 affects the cell cycle by altering the expression of Cyclin D1, which, in turn, affects the incidence and growth of malignancies and promotes cell differentiation. Previous reports indicate that APR3 is located in lysosomal membranes, where it contributes to lysosomal activity and participates in autophagy. While further research is required to determine the precise role and molecular mechanisms of APR3, earlier studies have laid the groundwork for APR3 research. There is growing evidence supporting the significance of APR3 in oncology. Therefore, this review aims to examine the current state of knowledge on the role of the newly discovered APR3 in tumorigenesis and to generate fresh insights and suggestions for future research.
Collapse
Affiliation(s)
- Ping Zhang
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital(Affiliated People's Hospital), Hangzhou Medical College, 310014, Hangzhou, Zhejiang, China
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Department of Clinical Research Center, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, 310006, Hangzhou, Zhejiang, China
- School of Pharmacy, Hangzhou Medical College, 310000, Hangzhou, Zhejiang, China
| | - Chaoting Zhou
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital(Affiliated People's Hospital), Hangzhou Medical College, 310014, Hangzhou, Zhejiang, China
| | - Qiangan Jing
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital(Affiliated People's Hospital), Hangzhou Medical College, 310014, Hangzhou, Zhejiang, China
| | - Yan Gao
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital(Affiliated People's Hospital), Hangzhou Medical College, 310014, Hangzhou, Zhejiang, China
- School of Pharmacy, Hangzhou Medical College, 310000, Hangzhou, Zhejiang, China
| | - Lei Yang
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital(Affiliated People's Hospital), Hangzhou Medical College, 310014, Hangzhou, Zhejiang, China
| | - Yanchun Li
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Department of Clinical Research Center, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, 310006, Hangzhou, Zhejiang, China
| | - Jing Du
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital(Affiliated People's Hospital), Hangzhou Medical College, 310014, Hangzhou, Zhejiang, China.
| | - Xiangmin Tong
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital(Affiliated People's Hospital), Hangzhou Medical College, 310014, Hangzhou, Zhejiang, China.
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Department of Clinical Research Center, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, 310006, Hangzhou, Zhejiang, China.
| | - Ying Wang
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital(Affiliated People's Hospital), Hangzhou Medical College, 310014, Hangzhou, Zhejiang, China.
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Department of Clinical Research Center, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, 310006, Hangzhou, Zhejiang, China.
- Department of Clinical Research Center, Luqiao Second People's Hospital, 317200, Taizhou, Zhejiang, China.
| |
Collapse
|
3
|
Nana FA, Lamberts V, Hoton D, Stanciu CP, Lecocq M, Carlier FM, Duplaquet F, Pirard L, Pilette C, Bihin B, Ocak S. Restin protein expression in non-small cell lung cancer. Thorac Cancer 2023; 14:2302-2309. [PMID: 37365889 PMCID: PMC10423652 DOI: 10.1111/1759-7714.15019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 06/13/2023] [Accepted: 06/14/2023] [Indexed: 06/28/2023] Open
Abstract
BACKGROUND Restin is a member of the melanoma-associated antigen (MAGE) superfamily. Its expression has been reported to be up- or downregulated in cancer. Preclinical data suggest it is a tumor suppressor. In this study, we aimed to evaluate restin expression and prognostic value in non-small cell lung cancer (NSCLC). METHODS Restin expression was analyzed by immunohistochemistry in three tissue microarrays consisting of formalin-fixed/paraffin-embedded NSCLC specimens from 113 patients, represented in triplicate. Restin staining H-score was the result of the staining intensity (0-no, 1-weak, 2-moderate, and 3-strong) multiplied by the percentage of stained tumor cells; it was defined as low if 1-100, moderate if 101-200, and strong if 201-300. Haverage-score was the average H-score in the triplicate. Restin Haverage-scores were tested for correlations with clinical and pathological characteristics and patient outcome. RESULTS Restin expression was localized to the cytoplasm, with nuclear enhancement, of 112/113 (99.1%) NSCLCs. Restin Haverage-scores were 0 in 1/113 (0.88%), low in 15/113 (13.3%), moderate in 48/113 (42.5%), and strong in 49/113 (43.4%) NSCLCs. Restin Haverage-scores did not correlate with NSCLC histological subtype, disease stage, recurrence/progression-free, or overall survival. CONCLUSION Restin is moderately to strongly expressed in the majority of NSCLC tumors but its expression has no prognostic value in patients with NSCLC.
Collapse
Affiliation(s)
- Frank Aboubakar Nana
- Division of PulmonologyCliniques Universitaires Saint‐LucBrusselsBelgium
- Pole of Pneumology, ENT, and Dermatology (PNEU), Institut de Recherche Expérimentale et Clinique (IREC)Université catholique de Louvain (UCLouvain)BrusselsBelgium
| | | | - Delphine Hoton
- Department of PathologyCliniques Universitaires Saint‐LucBrusselsBelgium
| | | | - Marylène Lecocq
- Pole of Pneumology, ENT, and Dermatology (PNEU), Institut de Recherche Expérimentale et Clinique (IREC)Université catholique de Louvain (UCLouvain)BrusselsBelgium
| | - François M. Carlier
- Pole of Pneumology, ENT, and Dermatology (PNEU), Institut de Recherche Expérimentale et Clinique (IREC)Université catholique de Louvain (UCLouvain)BrusselsBelgium
- Division of Pulmonology, CHU UCL NamurYvoirBelgium
| | | | | | - Charles Pilette
- Division of PulmonologyCliniques Universitaires Saint‐LucBrusselsBelgium
- Pole of Pneumology, ENT, and Dermatology (PNEU), Institut de Recherche Expérimentale et Clinique (IREC)Université catholique de Louvain (UCLouvain)BrusselsBelgium
| | - Benoît Bihin
- Scientific Support Unit, CHU UCL NamurYvoirBelgium
| | - Sebahat Ocak
- Pole of Pneumology, ENT, and Dermatology (PNEU), Institut de Recherche Expérimentale et Clinique (IREC)Université catholique de Louvain (UCLouvain)BrusselsBelgium
- Division of Pulmonology, CHU UCL NamurYvoirBelgium
| |
Collapse
|
4
|
Zhang L, Yang C, Huang Y, Huang H, Yuan X, Zhang P, Ye C, Wei M, Wang Y, Luo X, Luo J. Cardamonin inhibits the growth of human osteosarcoma cells through activating P38 and JNK signaling pathway. Biomed Pharmacother 2021; 134:111155. [PMID: 33370628 DOI: 10.1016/j.biopha.2020.111155] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 12/11/2020] [Accepted: 12/14/2020] [Indexed: 02/08/2023] Open
Abstract
Osteosarcoma (OS) is the most common type of bone malignant tumors. Clinical commonly used therapeutic drugs of OS treatment are prone to toxic and side effects, so it is very urgent to develop new drugs with low toxicity and low side effects. As a Chinese herbal medicine, Cardamonin (CAR) (C16H14O4) has inhibitory effects in various tumors. In the present study, we investigated the effects of CAR on OS cells in vitro and in vivo. We found that CAR inhibited cell proliferation, reduced migration, decreased invasion, and induced G2 / M arrest of OS cells. Notably, we demonstrated that CAR had no obvious effect on proliferation and apoptosis of normal cells. Besides, CAR repressed tumor growth of OS cells in xenograft mouse model. Mechanically, we found that CAR increased the phosphorylation level of P38 and JNK. In summary, our research validates that CAR may inhibit the proliferation, migration, and invasion of OS and promote apoptosis possibly by activating P38 and JNK Mitogen-activated protein kinase (MAPK) signaling pathway.
Collapse
Affiliation(s)
- Lulu Zhang
- School of Laboratory Medicine, Chongqing Medical University, Chongqing, China.
| | - Chunmei Yang
- School of Laboratory Medicine, Chongqing Medical University, Chongqing, China.
| | - Yanran Huang
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| | - Huakun Huang
- School of Laboratory Medicine, Chongqing Medical University, Chongqing, China.
| | - Xiaohui Yuan
- School of Laboratory Medicine, Chongqing Medical University, Chongqing, China.
| | - Ping Zhang
- School of Laboratory Medicine, Chongqing Medical University, Chongqing, China.
| | - Caihong Ye
- School of Laboratory Medicine, Chongqing Medical University, Chongqing, China.
| | - Mengqi Wei
- School of Laboratory Medicine, Chongqing Medical University, Chongqing, China.
| | - Yuping Wang
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| | - Xiaoji Luo
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| | - Jinyong Luo
- School of Laboratory Medicine, Chongqing Medical University, Chongqing, China.
| |
Collapse
|
5
|
Kasemsuk T, Saehlim N, Arsakhant P, Sittithumcharee G, Okada S, Saeeng R. A novel synthetic acanthoic acid analogues and their cytotoxic activity in cholangiocarcinoma cells. Bioorg Med Chem 2021; 29:115886. [PMID: 33290909 DOI: 10.1016/j.bmc.2020.115886] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2020] [Revised: 11/01/2020] [Accepted: 11/07/2020] [Indexed: 12/15/2022]
Abstract
A novel series of acanthoic acid analogues containing triazole moiety were synthesized through esterification and CuAAC reaction. Evaluation of their biological activities against four cell lines of cholangiocarcinoma cells showed that 3d exhibited the strongest activity with an IC50 value of 18 µM against KKU-213 cell line, which was 8 fold more potent than acanthoic acid. Interestingly, the triazole ring and nitro group on benzyl ring play very significant role in cytotoxic activity. The computational studies revealed that 3d occupies the binding energy of -12.7 and -10.8 kcal/mol with CDK-2 and EGFR protein kinases, respectively. This result might provide a beginning for the development of acanthoic acid analogues as an anticancer agent.
Collapse
Affiliation(s)
- Teerapich Kasemsuk
- Department of Chemistry, Faculty of Science and Technology, Rambhai Barni Rajabhat University, Chanthaburi 22000, Thailand
| | - Natthiya Saehlim
- Department of Chemistry and Center for Innovation in Chemistry, Faculty of Science, Burapha University, Chonburi 20131, Thailand
| | - Patcharee Arsakhant
- Department of Chemistry and Center for Innovation in Chemistry, Faculty of Science, Burapha University, Chonburi 20131, Thailand
| | - Gunya Sittithumcharee
- Division of Hematopoiesis, Joint Research Center for Human Retrovirus Infection & Graduate School of Medical Sciences, Kumamoto University, Japan
| | - Seiji Okada
- Division of Hematopoiesis, Joint Research Center for Human Retrovirus Infection & Graduate School of Medical Sciences, Kumamoto University, Japan
| | - Rungnapha Saeeng
- Department of Chemistry and Center for Innovation in Chemistry, Faculty of Science, Burapha University, Chonburi 20131, Thailand; The Research Unit in Synthetic Compounds and Synthetic Analogues from Natural Product for Drug Discovery (RSND), Burapha University, Chonburi 20131, Thailand.
| |
Collapse
|
6
|
Arora M, Kumari S, Singh J, Chopra A, Chauhan SS. Downregulation of Brain Enriched Type 2 MAGEs Is Associated With Immune Infiltration and Poor Prognosis in Glioma. Front Oncol 2020; 10:573378. [PMID: 33425727 PMCID: PMC7787151 DOI: 10.3389/fonc.2020.573378] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 11/10/2020] [Indexed: 12/15/2022] Open
Abstract
Melanoma associated antigen (MAGE) is an extensively studied family of tumor-associated genes that share a common MAGE homology domain (MHD). Based upon their expression pattern, MAGE genes have been broadly classified into type 1 MAGEs (T1Ms) and type 2 MAGEs (T2Ms) categories. Interestingly, several T2Ms are highly expressed in the brain and involved in the regulation of neuronal development, differentiation, and survival. Available literature suggests possible tumor suppressor functions of a few T2Ms, while information available about their expression, regulation, and clinical significance in glioma is scanty. This prompted us to perform a comprehensive analysis of T2M expression in glioma. Gene expression data from glioma datasets: Oncomine, TCGA, and REMBRANDT study, were used to assess the mRNA expression of T2M genes (MAGED1, MAGED2, MAGED3, MAGED4, MAGED4B, MAGEE1, MAGEE2, MAGEF1, MAGEH1, MAGEL2, NSMCE3, and NDN), and their association with clinical characteristics and composition of the tumor microenvironment. Further, mutation, copy number alteration, and DNA methylation data from TCGA were assessed for determining potential mechanisms of T2Ms expression in glioma. Expression analysis revealed overexpression of MAGED subfamily genes in glioma, while other genes of this family exhibited reduced expression in advanced grades of this malignancy. Further, the expression of T2Ms exhibited varying extent of positive correlations with each other. Amongst downregulated T2Ms, MAGEH1 expression exhibited negative correlations with DNA methylation. Additionally, genes associated with MAGEH1 were enriched in Myc and Hedgehog signaling. Furthermore, T2Ms downregulation was associated with immune infiltration in glioma tissues and poor overall survival of glioma patients. In multivariate Cox regression analysis, MAGEH1 emerged as an independent prognosticator in lower grade glioma. Conclusively, these results suggest that expression of T2Ms is associated with important clinical and molecular features in glioma. Mechanistic studies may further provide novel insights into their role in glioma progression.
Collapse
Affiliation(s)
- Mohit Arora
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Sarita Kumari
- Laboratory Oncology Unit, Dr. B. R. Ambedkar Institute Rotary Cancer Hospital (Dr. BRA-IRCH), All India Institute of Medical Sciences, New Delhi, India
| | - Jay Singh
- Laboratory Oncology Unit, Dr. B. R. Ambedkar Institute Rotary Cancer Hospital (Dr. BRA-IRCH), All India Institute of Medical Sciences, New Delhi, India
| | - Anita Chopra
- Laboratory Oncology Unit, Dr. B. R. Ambedkar Institute Rotary Cancer Hospital (Dr. BRA-IRCH), All India Institute of Medical Sciences, New Delhi, India
| | - Shyam S Chauhan
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
7
|
Activity-Based Protein Profiling Reveals Potential Dasatinib Targets in Gastric Cancer. Int J Mol Sci 2020; 21:ijms21239276. [PMID: 33291786 PMCID: PMC7729964 DOI: 10.3390/ijms21239276] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 11/30/2020] [Accepted: 12/02/2020] [Indexed: 12/28/2022] Open
Abstract
Dasatinib is a multi-target kinase inhibitor, whose targets include BCR-ABL, SRC family kinases, and various cancer kinases. The elevated SRC activity in gastric cancer (GC) has prompted the need for the therapeutic application of dasatinib in GC. We observed that the efficacy of dasatinib varied with the GC cell lines. The differential effect of dasatinib was not correlated with the basal SRC activity of each cell line. Moreover, the GC cell lines showing the strong antitumor effects of dasatinib were refractory to other SRC inhibitors, i.e., bosutinib and saracatinib, suggesting that unexpected dasatinib’s targets could exist. To profile the targets of dasatinib in GC, we performed activity-based protein profiling (ABPP) via mass spectrometry using a desthiobiotin-ATP probe. We identified 29 and 18 kinases as potential targets in dasatinib-sensitive (SNU-216, MKN-1) and -resistant (SNU-484, SNU-601) cell lines, respectively. The protein–protein interaction mapping of the differential drug targets in dasatinib-sensitive and -resistant GC using the STRING database suggested that dasatinib could target cellular energy homeostasis in the drug-sensitive GC. RNAi screening for identified targets indicated p90RSK could be a novel dasatinib target, which is important for maintaining the viability and motility of GC cells. Further functional validation of dasatinib off-target actions will provide more effective therapeutic options for GC.
Collapse
|
8
|
Anjitha R, Antony A, Shilpa O, Anupama KP, Mallikarjunaiah S, Gurushankara HP. Malathion induced cancer-linked gene expression in human lymphocytes. ENVIRONMENTAL RESEARCH 2020; 182:109131. [PMID: 32069766 DOI: 10.1016/j.envres.2020.109131] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 01/08/2020] [Accepted: 01/09/2020] [Indexed: 06/10/2023]
Abstract
BACKGROUND Malathion is the most widely used organophosphate pesticide in agriculture. Increasing cancer incidence in agricultural workers and their children links to the exposure of malathion. Identification of genes involved in the process of carcinogenesis is essential for exploring the role of malathion. The alteration in gene expression by malathion in human lymphocytes has not been explored yet, although hematological malignancies are rampant in humans. OBJECTIVE This study investigates the malathion induced expression of cancer associated genes in human lymphocytes. METHODS Human lymphocyte viability and colony-forming ability were analyzed in malathion treated and control groups. Gene expression profile in control and malathion treated human lymphocytes were performed using a microarray platform. The genes which have significant functions and those involved in different pathways were analyzed using the DAVID database. Differential gene expression upon malathion exposure was validated by quantitative real-time (qRT)-PCR. RESULTS Malathion caused a concentration-dependent reduction in human lymphocyte viability. At low concentration (50 μg/mL) of malathion treatment, human lymphocytes were viable indicating that low concentration of malathion is not cytotoxic and induces the colony formation. Total of 659 genes (15%) were up regulated and 3729 genes (85%) were down regulated in malathion treated human lymphocytes. About 57 cancer associated genes related to the growth and differentiation of B and T cells, immunoglobulin production, haematopoiesis, tumor suppression, oncogenes and signal transduction pathways like MAPK and RAS were induced by malathion. CONCLUSION This study evidences the carcinogenic nature of malathion. Low concentration of this pesticide is not cytotoxic and induces differentially regulated genes in human lymphocytes, which are involved in the initiation, progression, and pathogenesis of cancer.
Collapse
Affiliation(s)
- Ramakrishnan Anjitha
- Department of Zoology, School of Biological Sciences, Central University of Kerala, Periya, 671 320, Kasaragod, Kerala, India
| | - Anet Antony
- Department of Zoology, School of Biological Sciences, Central University of Kerala, Periya, 671 320, Kasaragod, Kerala, India
| | - Olakkaran Shilpa
- Department of Zoology, School of Biological Sciences, Central University of Kerala, Periya, 671 320, Kasaragod, Kerala, India
| | - Kizhakke P Anupama
- Department of Zoology, School of Biological Sciences, Central University of Kerala, Periya, 671 320, Kasaragod, Kerala, India
| | - Shanthala Mallikarjunaiah
- Center for Applied Genetics, Department of Studies in Zoology, Bangalore University, Jnanabharathi, Bengaluru, 560 056, Karnataka, India
| | - Hunasanahally P Gurushankara
- Department of Zoology, School of Biological Sciences, Central University of Kerala, Periya, 671 320, Kasaragod, Kerala, India.
| |
Collapse
|
9
|
Shankar EPS, Bahulayan D. Chemistry, chemical biology and photophysics of certain new chromene–triazole–coumarin triads as fluorescent inhibitors of CDK2 and CDK4 induced cancers. NEW J CHEM 2019. [DOI: 10.1039/c9nj02924a] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The chromene–triazole–coumarin triads synthesized through the MCR-Click strategy possess intense solution state fluorescence, intense solid-state fluorescence and CDK2/CDK4 targeted cytotoxic activity against human cervical cancer cells (HeLa).
Collapse
|
10
|
He Y, Fan Q, Cai T, Huang W, Xie X, Wen Y, Shi Z. Molecular mechanisms of the action of Arctigenin in cancer. Biomed Pharmacother 2018; 108:403-407. [PMID: 30236849 DOI: 10.1016/j.biopha.2018.08.158] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2018] [Revised: 08/30/2018] [Accepted: 08/31/2018] [Indexed: 11/25/2022] Open
Abstract
Since antediluvian times, the scientific community has realized that natural compounds exhibit enormous potential for the treatment of terrible diseases, such as cancer. Despite a variety of effective bioactive molecules, effective therapies still need to be developed to treat cancer. Hence, it is necessary to study the interactions of natural molecules with their cellular targets. Arctigenin (ATG), a natural lignan compound extracted from Arctium lappa, inhibits the growth of various cancer cells, such as those of the stomach, lungs, liver, and colon, as well as leukocytes, and regulates numerous intracellular activities, such as antioxidative, anti-inflammatory, and anticancer activities. The intention of this paper is to summarize and generally analyse the molecular pathways that are involved in the anticancer effects of ATG. In addition, the interactions of ATG with other drugs are also highlighted in this paper.
Collapse
Affiliation(s)
- Yinghua He
- Department of Pharmacy, The First Affiliated Hospital of Zhejiang Chinese Medicine University, Xiasha District, Hangzhou, Zhejiang 310018, China; Department of Pharmacy, Zhejiang International Exchange Center of Clinical Traditional Chinese Medicine, Xiasha District, Hangzhou, Zhejiang 310018, China; Department of Pharmacy, Zhejiang Provincial Hospital of Traditional Chinese Medicine, Xiasha District, Hangzhou, Zhejiang 310018, China
| | - Qiaomei Fan
- Department of Pharmacy, The First Affiliated Hospital of Zhejiang Chinese Medicine University, Xiasha District, Hangzhou, Zhejiang 310018, China; Department of Pharmacy, Zhejiang International Exchange Center of Clinical Traditional Chinese Medicine, Xiasha District, Hangzhou, Zhejiang 310018, China; Department of Pharmacy, Zhejiang Provincial Hospital of Traditional Chinese Medicine, Xiasha District, Hangzhou, Zhejiang 310018, China
| | - Tiantian Cai
- Department of Pharmacy, The First Affiliated Hospital of Zhejiang Chinese Medicine University, Xiasha District, Hangzhou, Zhejiang 310018, China; Department of Pharmacy, Zhejiang International Exchange Center of Clinical Traditional Chinese Medicine, Xiasha District, Hangzhou, Zhejiang 310018, China; Department of Pharmacy, Zhejiang Provincial Hospital of Traditional Chinese Medicine, Xiasha District, Hangzhou, Zhejiang 310018, China
| | - Wei Huang
- Department of Pharmacy, The First Affiliated Hospital of Zhejiang Chinese Medicine University, Xiasha District, Hangzhou, Zhejiang 310018, China; Department of Pharmacy, Zhejiang International Exchange Center of Clinical Traditional Chinese Medicine, Xiasha District, Hangzhou, Zhejiang 310018, China; Department of Pharmacy, Zhejiang Provincial Hospital of Traditional Chinese Medicine, Xiasha District, Hangzhou, Zhejiang 310018, China
| | - Xianze Xie
- Department of Pharmacy, The First Affiliated Hospital of Zhejiang Chinese Medicine University, Xiasha District, Hangzhou, Zhejiang 310018, China; Department of Pharmacy, Zhejiang International Exchange Center of Clinical Traditional Chinese Medicine, Xiasha District, Hangzhou, Zhejiang 310018, China; Department of Pharmacy, Zhejiang Provincial Hospital of Traditional Chinese Medicine, Xiasha District, Hangzhou, Zhejiang 310018, China
| | - Yayun Wen
- Department of Pharmacy, The First Affiliated Hospital of Zhejiang Chinese Medicine University, Xiasha District, Hangzhou, Zhejiang 310018, China; Department of Pharmacy, Zhejiang International Exchange Center of Clinical Traditional Chinese Medicine, Xiasha District, Hangzhou, Zhejiang 310018, China; Department of Pharmacy, Zhejiang Provincial Hospital of Traditional Chinese Medicine, Xiasha District, Hangzhou, Zhejiang 310018, China
| | - Zheng Shi
- Department of Pharmacy, The First Affiliated Hospital of Zhejiang Chinese Medicine University, Xiasha District, Hangzhou, Zhejiang 310018, China; Department of Pharmacy, Zhejiang International Exchange Center of Clinical Traditional Chinese Medicine, Xiasha District, Hangzhou, Zhejiang 310018, China; Department of Pharmacy, Zhejiang Provincial Hospital of Traditional Chinese Medicine, Xiasha District, Hangzhou, Zhejiang 310018, China.
| |
Collapse
|
11
|
Wu F, Shi X, Zhang R, Tian Y, Wang X, Wei C, Li D, Li X, Kong X, Liu Y, Guo W, Guo Y, Zhou H. Regulation of proliferation and cell cycle by protein regulator of cytokinesis 1 in oral squamous cell carcinoma. Cell Death Dis 2018; 9:564. [PMID: 29752448 PMCID: PMC5948203 DOI: 10.1038/s41419-018-0618-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2017] [Accepted: 04/24/2018] [Indexed: 02/05/2023]
Abstract
Protein regulator of cytokinesis 1 (PRC1), a microtubule-associated protein, has emerged as a critical regulator of proliferation and apoptosis, acting predominantly in numerous tumors. However, its function in oral squamous cell carcinoma (OSCC) is still unknown. To establish the roles of PRC1 in OSCC, 95 oral clinical samples (54 OSCC, 24 oral leukoplakia [OLK], and 17 normal oral mucosa) and seven oral cell lines (6 OSCC and 1 normal oral cell lines) were analyzed using a series of molecular and genomic assays both in vivo and in vitro were conducted in this study. Herein, we provide evidence demonstrating that expression of PRC1 closely correlates with the degree of epithelial dysplasia in OLK (n = 24) (p < 0.001), and the poor differentiation, large tumor volume, lymph node metastasis, and high-clinical stage in OSCC (n = 54) (p < 0.05), illustrating that PRC1 has a promotive influence on tumor progression in OSCC. Simultaneously, we observed that PRC1 knockdown in OSCC cell lines caused G2/M phase arrest (p < 0.05), inhibited cell proliferation in vitro (p < 0.05) and tumor growth in vivo (p < 0.001). Furthermore, the effects of PRC1 on the regulation of proliferation and cell cycle transition in OSCC samples were mediated by p53. The p53/PRC1/EGFR signaling pathway was found to be implicated in the tumor progression of OSCC. Based on our data, we demonstrate that PRC1 is a key factor in regulating proliferation and the cell cycle, pointing to the potential benefits of PRC1-targeted therapies for OSCC.
Collapse
Affiliation(s)
- Fanglong Wu
- State Key Laboratory of Oral Diseases, Department of Oral Medicine, West China Hospital of Stomatology, Sichuan University, 610041, Chengdu, Sichuan, China
| | - Xueke Shi
- State Key Laboratory of Oral Diseases, Department of Oral Medicine, West China Hospital of Stomatology, Sichuan University, 610041, Chengdu, Sichuan, China
| | - Rui Zhang
- Department of Preventive Dentistry, West China Hospital of Stomatology, Sichuan University, 610041, Chengdu, Sichuan, China
| | - Yuan Tian
- State Key Laboratory of Oral Diseases, Department of Oral Medicine, West China Hospital of Stomatology, Sichuan University, 610041, Chengdu, Sichuan, China
| | - Xiangjian Wang
- State Key Laboratory of Oral Diseases, Department of Oral Medicine, West China Hospital of Stomatology, Sichuan University, 610041, Chengdu, Sichuan, China
| | - Changlei Wei
- State Key Laboratory of Oral Diseases, Department of Oral Medicine, West China Hospital of Stomatology, Sichuan University, 610041, Chengdu, Sichuan, China
| | - Duo Li
- State Key Laboratory of Oral Diseases, Department of Oral Medicine, West China Hospital of Stomatology, Sichuan University, 610041, Chengdu, Sichuan, China
| | - Xiaoyu Li
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, 610041, Chengdu, Sichuan, China
| | - Xiangli Kong
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, 610041, Chengdu, Sichuan, China
| | - Yurong Liu
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, 610041, Chengdu, Sichuan, China
| | - Weihua Guo
- Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, 610041, Chengdu, Sichuan, China.
| | - Yiqing Guo
- State Key Laboratory of Oral Diseases, Department of Oral Medicine, West China Hospital of Stomatology, Sichuan University, 610041, Chengdu, Sichuan, China. .,Department of Stomatology, The Affiliated Hospital of Qingdao University, 266003, Qingdao, Shandong, China.
| | - Hongmei Zhou
- State Key Laboratory of Oral Diseases, Department of Oral Medicine, West China Hospital of Stomatology, Sichuan University, 610041, Chengdu, Sichuan, China.
| |
Collapse
|
12
|
Wang PC, Hu ZQ, Zhou SL, Zhan H, Zhou ZJ, Luo CB, Huang XW. Downregulation of MAGE family member H1 enhances hepatocellular carcinoma progression and serves as a biomarker for patient prognosis. Future Oncol 2018; 14:1177-1186. [PMID: 29316827 DOI: 10.2217/fon-2017-0672] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
AIM The MAGE family member H1 (MAGEH1) belongs to melanoma-associated antigen (MAGE) superfamily. The role of MAGEH1 in hepatocellular carcinoma (HCC) is largely undefined. MATERIALS & METHODS We used quantitative reverse transcription PCR and immunohistochemistry to detect MAGEH1 expression in HCC tissues. CCK-8 assay, wound healing migration assay and Transwell Matrigel invasion assay were used to measure HCC cell proliferation, migration and invasion ability. RESULTS MAGEH1 expression was downregulated in HCC tumor tissues compared with adjacent normal liver tissues and in samples from patients with tumor recurrence. MAGEH1 reduced HCC cell proliferation, migration and invasion ability. Low MAGEH1 expression was significantly correlated with poor prognosis in HCC patients. CONCLUSION MAGEH1 may serve as a potential biomarker and a new prognostic factor for HCC.
Collapse
Affiliation(s)
- Peng-Cheng Wang
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis & Cancer Invasion, Fudan University, Ministry of Education, Shanghai 200032, PR China
| | - Zhi-Qiang Hu
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis & Cancer Invasion, Fudan University, Ministry of Education, Shanghai 200032, PR China
| | - Shao-Lai Zhou
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis & Cancer Invasion, Fudan University, Ministry of Education, Shanghai 200032, PR China
| | - Hao Zhan
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis & Cancer Invasion, Fudan University, Ministry of Education, Shanghai 200032, PR China
| | - Zheng-Jun Zhou
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis & Cancer Invasion, Fudan University, Ministry of Education, Shanghai 200032, PR China
| | - Chu-Bin Luo
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis & Cancer Invasion, Fudan University, Ministry of Education, Shanghai 200032, PR China
| | - Xiao-Wu Huang
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis & Cancer Invasion, Fudan University, Ministry of Education, Shanghai 200032, PR China
| |
Collapse
|
13
|
Han YH, Kee JY, Kim DS, Mun JG, Park SH, Kim YJ, Um JY, Hong SH. Arctii Fructus Inhibits Colorectal Cancer Cell Proliferation and MMPs Mediated Invasion via AMPK. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2017; 45:1309-1325. [PMID: 28830210 DOI: 10.1142/s0192415x17500720] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Although Arctii Fructus (AF) has been shown to have various pharmacological effects, there have been no studies concerning the inhibitory effects of AF on the metastatic properties of colorectal cancer (CRC). The aim of this study was to investigate whether AF could suppress CRC progression by inhibiting cell growth, epithelial-mesenchymal transition (EMT), migration, and the invasion ability of CRC cells. AF decreased proliferation of CRC cells by inducing cell cycle arrest and apoptosis via extrinsic and intrinsic apoptotic pathways. Regarding metastatic properties, AF inhibited EMT by increasing the expression of the epithelial marker, E-cadherin, and decreasing the expression of the mesenchymal marker, N-cadherin, in CT26 cells. Moreover, AF decreased the migration and invasion of CT26 cells by inhibiting matrix metalloproteinase-2 (MMP-2) and MMP-9 activity. We confirmed that the decreased invasion ability and MMP-9 activity by AF treatment involved AMP-activated protein kinase (AMPK) activation. Collectively, this study demonstrates that AF inhibits the proliferation and metastatic properties of CRC cells.
Collapse
Affiliation(s)
- Yo-Han Han
- * Department of Oriental Pharmacy, College of Pharmacy, Wonkwang-Oriental Medicines Research Institute, Wonkwang University, 460 Iksandae-ro, Iksan, Jeonbuk 54538, Republic of Korea
| | - Ji-Ye Kee
- * Department of Oriental Pharmacy, College of Pharmacy, Wonkwang-Oriental Medicines Research Institute, Wonkwang University, 460 Iksandae-ro, Iksan, Jeonbuk 54538, Republic of Korea
| | - Dae-Seung Kim
- * Department of Oriental Pharmacy, College of Pharmacy, Wonkwang-Oriental Medicines Research Institute, Wonkwang University, 460 Iksandae-ro, Iksan, Jeonbuk 54538, Republic of Korea
| | - Jeong-Geon Mun
- * Department of Oriental Pharmacy, College of Pharmacy, Wonkwang-Oriental Medicines Research Institute, Wonkwang University, 460 Iksandae-ro, Iksan, Jeonbuk 54538, Republic of Korea
| | - Seong-Hwan Park
- * Department of Oriental Pharmacy, College of Pharmacy, Wonkwang-Oriental Medicines Research Institute, Wonkwang University, 460 Iksandae-ro, Iksan, Jeonbuk 54538, Republic of Korea
| | - Yong Ju Kim
- † Department of Herbal Medicine Resources, College of Environmental and Bioresources Sciences, Chonbuk National University, 54596 Iksan, Republic of Korea
| | - Jae-Young Um
- ‡ Department of Pharmacology, College of Korean Medicine, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea
| | - Seung-Heon Hong
- * Department of Oriental Pharmacy, College of Pharmacy, Wonkwang-Oriental Medicines Research Institute, Wonkwang University, 460 Iksandae-ro, Iksan, Jeonbuk 54538, Republic of Korea
| |
Collapse
|
14
|
Wang J, Yang T, Xu G, Liu H, Ren C, Xie W, Wang M. Cyclin-Dependent Kinase 2 Promotes Tumor Proliferation and Induces Radio Resistance in Glioblastoma. Transl Oncol 2016; 9:548-556. [PMID: 27863310 PMCID: PMC5118617 DOI: 10.1016/j.tranon.2016.08.007] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2016] [Revised: 08/18/2016] [Accepted: 08/18/2016] [Indexed: 11/03/2022] Open
Abstract
Accumulating evidence indicates that CDK2 promotes hyperproliferation and is associated to poor prognosis in multiple cancer cells. However, the physiological role of CDK2 in GBM and the biological mechanism still remains unclear. In this study, we identified that CDK2 expression was significantly enriched in GBM tumors compared with normal brain. Additionally, CDK2 was functionally required for tumor proliferation and its expression was associated to poor prognosis in GBM patients. Mechanically, CDK2 induced radio resistance in GBM cells and CDK2 knock down increased cell apoptosis when combined with radiotherapy. Therapeutically, we found that CDK2 inhibitor attenuated tumor growth both in vitro and in vivo. Collectively, CDK2 promotes proliferation, induces radio resistance in GBM, and could become a therapeutic target for GBM.
Collapse
Affiliation(s)
- Jia Wang
- Department of Neurosurgery, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China; School of Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Tong Yang
- Department of Neurosurgery, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China; School of Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Gaofeng Xu
- Department of Neurosurgery, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Hao Liu
- Department of Neurosurgery, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Chunying Ren
- Department of Neurosurgery, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Wanfu Xie
- Department of Neurosurgery, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China.
| | - Maode Wang
- Department of Neurosurgery, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China.
| |
Collapse
|
15
|
Arctigenin Inhibits Lung Metastasis of Colorectal Cancer by Regulating Cell Viability and Metastatic Phenotypes. Molecules 2016; 21:molecules21091135. [PMID: 27618887 PMCID: PMC6272973 DOI: 10.3390/molecules21091135] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Revised: 08/24/2016] [Accepted: 08/24/2016] [Indexed: 02/03/2023] Open
Abstract
Arctigenin (ARC) has been shown to have an anti-cancer effect in various cell types and tissues. However, there have been no studies concerning metastatic colorectal cancer (CRC). In this study, we investigated the anti-metastatic properties of ARC on colorectal metastasis and present a potential candidate drug. ARC induced cell cycle arrest and apoptosis in CT26 cells through the intrinsic apoptotic pathway via MAPKs signaling. In several metastatic phenotypes, ARC controlled epithelial-mesenchymal transition (EMT) through increasing the expression of epithelial marker E-cadherin and decreasing the expressions of mesenchymal markers; N-cadherin, vimentin, β-catenin, and Snail. Moreover, ARC inhibited migration and invasion through reducing of matrix metalloproteinase-2 (MMP-2) and MMP-9 expressions. In an experimental metastasis model, ARC significantly inhibited lung metastasis of CT26 cells. Taken together, our study demonstrates the inhibitory effects of ARC on colorectal metastasis.
Collapse
|