1
|
Nicolaescu OE, Ionescu C, Samide A, Tigae C, Spînu CI, Oprea B. Advancements in Cyclodextrin Complexes with Bioactive Secondary Metabolites and Their Pharmaceutical Applications. Pharmaceutics 2025; 17:506. [PMID: 40284503 PMCID: PMC12030412 DOI: 10.3390/pharmaceutics17040506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2025] [Revised: 04/09/2025] [Accepted: 04/09/2025] [Indexed: 04/29/2025] Open
Abstract
Cyclodextrins (CDs) have largely been investigated during the last decades for their outstanding properties, such as biocompatibility and biodegradability, with wide applications in the pharmaceutical field, among which the formation of inclusion complexes (ICs) with natural or synthetic lipophilic compounds. This review prioritizes the research of recent years (2022-2025), being focused on (1) systematization of the research of ICs based on the structure of the secondary metabolite, namely (i) polyphenols (PPs), (ii) terpenes and terpenoids (TTs), and (iii) alkaloids (Alks); (2) for each type of inclusion complex, the following aspects have been discussed: benefits of complexation, composite materials, and in vitro/in vivo and theoretical studies; and (3) pharmacokinetics and pharmacodynamics, risks, limitations, and perspectives of cyclodextrin inclusion complexes with secondary metabolites.
Collapse
Affiliation(s)
- Oana Elena Nicolaescu
- Department of Pharmaceutical Technique, Faculty of Pharmacy, University of Medicine and Pharmacy of Craiova, 2 Petru Rareş, 200349 Craiova, Dolj, Romania;
| | - Cătălina Ionescu
- Department of Chemistry, Faculty of Sciences, University of Craiova, 107i Calea București, 200144 Craiova, Dolj, Romania; (A.S.); (C.T.); (C.I.S.)
| | - Adriana Samide
- Department of Chemistry, Faculty of Sciences, University of Craiova, 107i Calea București, 200144 Craiova, Dolj, Romania; (A.S.); (C.T.); (C.I.S.)
| | - Cristian Tigae
- Department of Chemistry, Faculty of Sciences, University of Craiova, 107i Calea București, 200144 Craiova, Dolj, Romania; (A.S.); (C.T.); (C.I.S.)
| | - Cezar Ionuţ Spînu
- Department of Chemistry, Faculty of Sciences, University of Craiova, 107i Calea București, 200144 Craiova, Dolj, Romania; (A.S.); (C.T.); (C.I.S.)
| | - Bogdan Oprea
- Faculty of Medicine, University of Medicine and Pharmacy of Craiova, 2 Petru Rares, 200349 Craiova, Dolj, Romania;
| |
Collapse
|
2
|
Li Y, Zhou J, Liu L, Zhu C, Luo Z, Li N, Lyu P, Zhang J, Xie T, Ding Y, Xiao S. Association of SNPs in nAChRs genes, areca nut chewing and smoking, and their interaction with lung cancer in Hainan, China: a case control study. BMC Cancer 2025; 25:626. [PMID: 40197297 PMCID: PMC11974198 DOI: 10.1186/s12885-025-14020-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 03/25/2025] [Indexed: 04/10/2025] Open
Abstract
BACKGROUND Areca nut (AN) was classified as a carcinogen by the International Agency for Research on Cancer (IARC) of the WHO in 2003. AN has the same carcinogenic components as cigarettes, such as benzo[a]pyrene (B[a]P) and 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK), but its effects on interactions with genetic factors related to lung cancer have rarely been investigated. METHODS: Here, a propensity score-matched case‒control study was conducted in Hainan, which included 445 patients with lung cancer and 445 cancer-free controls. Then, the associations between single-nucleotide polymorphisms (SNPs) in the CHRNA5-CHRNA3-CHRNB4 gene cluster and their interaction effects with AN chewing and smoking on lung cancer were analyzed. In addition, we explored the associations among AN, cigarettes, and genes related to lung cancer using the Comparative Toxicogenomics Database (CTD). RESULTS The results indicate that the CHRNA3 rs938682 (A > G) GG genotype (OR = 0.669, 95% CI = 0.454 ~ 0.984, P = 0.042) can decrease the risk of lung cancer. The CHRNB4 rs7178270 (C > G) GG genotype (OR = 1.729, 95% CI = 1.168 ~ 2.571, P = 0.006) can increase the risk of lung cancer. The CHRNA5 rs17486278 CC genotype was associated with a high risk in males, smokers, and drinkers. The CHRNA3 rs938682 GG genotype was associated with a low risk in AN chewers. The CHRNB4 rs7178270 GG genotype was associated with high risk in drinkers and AN chewers. CHRNB4 rs7178270 and AN chewing have an interaction effect on lung cancer in Hainan. CONCLUSIONS This study is the first to elucidate the hidden impacts of AN on lung cancer and provides a key evidence regarding the interactive effects of AN and cigarettes with SNPs in nAChRs genes on lung cancer.
Collapse
Affiliation(s)
- Yixuan Li
- School of Public Health, Key Laboratory of Tropical Translational Medicine of Ministry of Education, Heinz Mehlhorn Academician Workstation, Hainan Medical University, Haikou, Hainan, 571199, People's Republic of China
| | - Jing Zhou
- School of Public Health, Key Laboratory of Tropical Translational Medicine of Ministry of Education, Heinz Mehlhorn Academician Workstation, Hainan Medical University, Haikou, Hainan, 571199, People's Republic of China
| | - Lirong Liu
- Department of Respiratory and Critical Care Medicine, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan, 570311, People's Republic of China
| | - Chaoyong Zhu
- Medical Examination Center of Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan, 570311, People's Republic of China
| | - Ziyue Luo
- School of Public Health, Key Laboratory of Tropical Translational Medicine of Ministry of Education, Heinz Mehlhorn Academician Workstation, Hainan Medical University, Haikou, Hainan, 571199, People's Republic of China
| | - Na Li
- School of Public Health, Key Laboratory of Tropical Translational Medicine of Ministry of Education, Heinz Mehlhorn Academician Workstation, Hainan Medical University, Haikou, Hainan, 571199, People's Republic of China
| | - Pengfei Lyu
- Department of Breast Surgery, The First Affiliated Hospital of Hainan Medical University, Haikou, Hainan, 570102, People's Republic of China
| | - Jing Zhang
- School of Public Health, Key Laboratory of Tropical Translational Medicine of Ministry of Education, Heinz Mehlhorn Academician Workstation, Hainan Medical University, Haikou, Hainan, 571199, People's Republic of China
| | - Tian Xie
- Department of Respiratory and Critical Care Medicine, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan, 570311, People's Republic of China
| | - Yipeng Ding
- Department of Respiratory and Critical Care Medicine, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan, 570311, People's Republic of China.
| | - Sha Xiao
- School of Public Health, Key Laboratory of Tropical Translational Medicine of Ministry of Education, Heinz Mehlhorn Academician Workstation, Hainan Medical University, Haikou, Hainan, 571199, People's Republic of China.
| |
Collapse
|
3
|
Wang W. Assessing the carcinogenic potential and molecular mechanisms of arecoline in human lungs: from in silico methods to in vitro validation. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 294:118108. [PMID: 40147173 DOI: 10.1016/j.ecoenv.2025.118108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Revised: 02/26/2025] [Accepted: 03/24/2025] [Indexed: 03/29/2025]
Abstract
OBJECTIVE Despite the globally recognized carcinogenic potential of arecoline, the primary active compound in areca nut, the molecular mechanisms underlying its role in lung adenocarcinoma (LUAD) have yet to be fully understood. This study aims to bridge this gap by integrating network toxicology, molecular docking and dynamics simulation, tumor bioinformatics, and in vitro assays to elucidate the molecular mechanisms through which arecoline contributes to LUAD development. METHODS We first utilized disease-related databases and compound databases to identify arecoline-targeted LUAD-relevant proteins and constructed an interaction network using Cytoscape to screen core proteins based on topological analysis. Subsequently, we performed molecular docking and dynamics simulation, along with surface plasmon resonance assay to examine and validate the interactions between arecoline and core proteins. Next, we utilized these proteins for functional enrichment analyses to explore their correlation with cancer. Ultimately, we detected the expression and prognosis of core genes and constructed a prognostic model to examine its relationship with immune infiltration and immunotherapy. RESULTS Arecoline targets 106 LUAD-relevant proteins, including 24 core proteins. The stable interactions of arecoline and core proteins (especially PTGS2) greatly support the carcinogenic toxicity of arecoline in human lungs. These target proteins influence the occurrence, progression, and immune infiltration of LUAD by participating in pathways related to cancer and immunity, thereby affecting the prognosis and immunotherapy of LUAD patients. CONCLUSION This study elucidates the molecular mechanism underlying arecoline-induced LUAD, introducing a novel approach for assessing food safety and presenting innovative and promising targets and strategies for cancer intervention and therapy.
Collapse
Affiliation(s)
- Wenwen Wang
- Translational Medicine Research Center, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, Zhejiang 310006, China.
| |
Collapse
|
4
|
Garg K, Kumar A, Kizhakkethil V, Kumar P, Singh S. Overlap in oncogenic and pro-inflammatory pathways associated with areca nut and nicotine exposure. CANCER PATHOGENESIS AND THERAPY 2024; 2:187-194. [PMID: 39027148 PMCID: PMC11252521 DOI: 10.1016/j.cpt.2023.09.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 09/08/2023] [Accepted: 09/14/2023] [Indexed: 07/20/2024]
Abstract
Background Betel nut/areca nut/Areca catechu is one of the most commonly used psychoactive substance, and is also a major preventable cause of cancer. Unlike other psychoactive substances, such as nicotine, the mechanisms underlying addiction to areca nuts and related oncogenesis remain elusive. Recent reports suggest a possible overlap in the mechanisms of action of nicotine and areca nuts in the human body. Thus, this study aimed to investigate the interactome of human proteins associated with areca nut exposure and the intricate similarities and differences in the effects of the two psychoactive substances on humans. Methods A list of proteins associated with areca nut use was obtained from the available literature using terms from Medical Subject Headings (MeSH). Protein-protein interaction (PPI) networks and functional enrichment were analyzed. The results obtained for both psychoactive substances were compared. Results Given the limited number of common proteins (36/226, 16%) in the two sets, a substantial overlap (612/1176 nodes, 52%) was observed in the PPI networks, as well as in Gene Ontology. Areca nuts mainly affect signaling pathways through three hub proteins (alpha serine/threonine-protein kinase, tumor protein 53, and interleukin-6), which are common to both psychoactive substances, as well as two unique hub proteins (epidermal growth factor receptor and master regulator of cell cycle entry and proliferative metabolism). Areca nut-related proteins are associated with unique pathways, such as extracellular matrix organization, lipid storage, and metabolism, which are not found in nicotine-associated proteins. Conclusions Areca nuts affect regulatory mechanisms, leading to systemic toxicity and oncogenesis. Areca nuts also affect unique pathways that can be studied as potential markers of exposure, as well as targets for anticancer therapeutic agents.
Collapse
Affiliation(s)
- Krati Garg
- Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology (IIT) Delhi, Delhi 110016, India
| | - Anuj Kumar
- Division of Molecular Biology, ICMR-National Institute of Cancer Prevention and Research (ICMR-NICPR), Indian Council of Medical Research, Noida, Uttar Pradesh 201301, India
| | - Vidisha Kizhakkethil
- Department of Biotechnology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu 632024, India
| | - Pramod Kumar
- Division of Molecular Biology, ICMR-National Institute of Cancer Prevention and Research (ICMR-NICPR), Indian Council of Medical Research, Noida, Uttar Pradesh 201301, India
| | - Shalini Singh
- ICMR-National Institute of Cancer Prevention & Research (ICMR-NICPR), Indian Council of Medical Research, Noida, Uttar Pradesh 201301, India
| |
Collapse
|
5
|
Oner M, Cheng PT, Wang HY, Chen MC, Lin H. Metformin alters dendrite development and synaptic plasticity in rat cortical neurons. Biochem Biophys Res Commun 2024; 710:149874. [PMID: 38581950 DOI: 10.1016/j.bbrc.2024.149874] [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: 03/08/2024] [Accepted: 03/29/2024] [Indexed: 04/08/2024]
Abstract
Synaptic plasticity is crucial as it dynamically molds the strength and connectivity of neural circuits, influencing learning, memory, and the development of neurological disorders. Metformin, a widely prescribed anti-diabetic medication, has been shown to readily cross the blood-brain barrier (BBB) and the placenta. However, its prolonged impact on neuronal morphology and functions remains underexplored. In this study, we investigated the influence of metformin on dendrite development and synaptic plasticity in embryonic brains and primary rat cortical neurons. Our findings reveal a negative modulation of dendrite development by metformin, as evidenced by altered dendritic arborization, impaired dendritic spine morphology and disruptions in synaptic plasticity, suggesting a potential link between metformin exposure and aberrations in neuronal connectivity. In addition, we extend our insights to the impact of maternal metformin exposure on embryonic brains, revealing a significant inhibition of dendrite development in E18.5 rat brains. In conclusion, this study adds to the expanding knowledge base on the non-metabolic effects of metformin, emphasizing the significance of assessing its potential influence on both neuronal structure and function. There is an urgent need for further investigations into the enduring impact of prolonged metformin administration on the structural and functional aspects of neurons.
Collapse
Affiliation(s)
- Muhammet Oner
- Department of Life Sciences, National Chung Hsing University, Taichung, 40227, Taiwan
| | - Pang-Ting Cheng
- Department of Life Sciences, National Chung Hsing University, Taichung, 40227, Taiwan
| | - Hsin-Yi Wang
- Department of Nuclear Medicine, Taichung Veterans General Hospital Taichung, 40705, Taiwan
| | - Mei-Chih Chen
- Translational Cell Therapy Center, Department of Medical Research, China Medical University Hospital, Taichung, 40447, Taiwan
| | - Ho Lin
- Department of Life Sciences, National Chung Hsing University, Taichung, 40227, Taiwan.
| |
Collapse
|
6
|
Sankar AP, Cho HM, Shin SU, Sneh T, Ramakrishnan S, Elledge C, Zhang Y, Das R, Gil-Henn H, Rosenblatt JD. Antibody-Drug Conjugate αEGFR-E-P125A Reduces Triple-negative Breast Cancer Vasculogenic Mimicry, Motility, and Metastasis through Inhibition of EGFR, Integrin, and FAK/STAT3 Signaling. CANCER RESEARCH COMMUNICATIONS 2024; 4:738-756. [PMID: 38315147 PMCID: PMC10926898 DOI: 10.1158/2767-9764.crc-23-0278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 11/14/2023] [Accepted: 01/24/2024] [Indexed: 02/07/2024]
Abstract
Primary tumor growth and metastasis in triple-negative breast cancer (TNBC) require supporting vasculature, which develop through a combination of endothelial angiogenesis and vasculogenic mimicry (VM), a process associated with aggressive metastatic behavior in which vascular-like structures are lined by tumor cells. We developed αEGFR-E-P125A, an antibody-endostatin fusion protein that delivers a dimeric, mutant endostatin (E-P125A) payload that inhibits TNBC angiogenesis and VM in vitro and in vivo. To characterize the mechanisms associated with induction and inhibition of VM, RNA sequencing (RNA-seq) of MDA-MB-231-4175 TNBC cells grown in a monolayer (two-dimensional) was compared with cells plated on Matrigel undergoing VM [three-dimensional (3D)]. We then compared RNA-seq between TNBC cells in 3D and cells in 3D with VM inhibited by αEGFR-E-P125A (EGFR-E-P125A). Gene set enrichment analysis demonstrated that VM induction activated the IL6-JAK-STAT3 and angiogenesis pathways, which were downregulated by αEGFR-E-P125A treatment.Correlative analysis of the phosphoproteome demonstrated decreased EGFR phosphorylation at Y1069, along with decreased phosphorylation of focal adhesion kinase Y397 and STAT3 Y705 sites downstream of α5β1 integrin. Suppression of phosphorylation events downstream of EGFR and α5β1 integrin demonstrated that αEGFR-E-P125A interferes with ligand-receptor activation, inhibits VM, and overcomes oncogenic signaling associated with EGFR and α5β1 integrin cross-talk. In vivo, αEGFR-E-P125A treatment decreased primary tumor growth and VM, reduced lung metastasis, and confirmed the inhibition of signaling events observed in vitro. Simultaneous inhibition of EGFR and α5β1 integrin signaling by αEGFR-E-P125A is a promising strategy for the inhibition of VM, tumor growth, motility, and metastasis in TNBC and other EGFR-overexpressing tumors. SIGNIFICANCE αEGFR-E-P125A reduces VM, angiogenesis, tumor growth, and metastasis by inhibiting EGFR and α5β1 integrin signaling, and is a promising therapeutic agent for TNBC treatment, used alone or in combination with chemotherapy.
Collapse
Affiliation(s)
- Ankita P. Sankar
- Department of Medicine, Division of Hematology, University of Miami Miller School of Medicine, Miami, Florida
- Sylvester Comprehensive Cancer Center, Miami, Florida
| | - Hyun-Mi Cho
- Department of Medicine, Division of Hematology, University of Miami Miller School of Medicine, Miami, Florida
- Sylvester Comprehensive Cancer Center, Miami, Florida
| | - Seung-Uon Shin
- Department of Medicine, Division of Hematology, University of Miami Miller School of Medicine, Miami, Florida
- Sylvester Comprehensive Cancer Center, Miami, Florida
| | - Tal Sneh
- The Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
| | - Sundaram Ramakrishnan
- Sylvester Comprehensive Cancer Center, Miami, Florida
- Dewitt Daughtry Department of Surgery, University of Miami Miller School of Medicine, Miami, Florida
| | - Christian Elledge
- Department of Medicine, Division of Hematology, University of Miami Miller School of Medicine, Miami, Florida
- Sylvester Comprehensive Cancer Center, Miami, Florida
| | - Yu Zhang
- Department of Medicine, Division of Hematology, University of Miami Miller School of Medicine, Miami, Florida
- Sylvester Comprehensive Cancer Center, Miami, Florida
| | - Rathin Das
- Synergys Biotherapeutics, Inc., Alamo, California
| | - Hava Gil-Henn
- The Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
| | - Joseph D. Rosenblatt
- Department of Medicine, Division of Hematology, University of Miami Miller School of Medicine, Miami, Florida
- Sylvester Comprehensive Cancer Center, Miami, Florida
| |
Collapse
|
7
|
Eslami SS, Jafari D, Ghotaslou A, Amoupour M, Asri Kojabad A, Jafari R, Mousazadeh N, Tarighi P, Sadeghizadeh M. Combined Treatment of Dendrosomal-Curcumin and Daunorubicin Synergistically Inhibit Cell Proliferation, Migration and Induce Apoptosis in A549 Lung Cancer Cells. Adv Pharm Bull 2023; 13:539-550. [PMID: 37646049 PMCID: PMC10460814 DOI: 10.34172/apb.2023.050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 05/13/2022] [Accepted: 07/01/2022] [Indexed: 09/01/2023] Open
Abstract
Purpose Chemotherapy drugs used to treat lung cancer are associated with drug resistance and severe side effects. There have been rising demands for new therapeutic candidates and novel approaches, including combination therapy. Here, we aimed to investigate the combinatorial effect of a dendrosomal formulation of curcumin (DNC) and daunorubicin (DNR) on the A549 lung cancer cell line. Methods We performed cytotoxicity, apoptosis, cell migration, colony-formation capacity, and gene expression analysis to interpret the mechanism of action for a combination of DNC and DNR on A549 cells. Results Our results revealed that the combination of DNC and DNR could synergistically inhibit the A549 cells' growth. This synergistic cytotoxicity was further approved by flow cytometry, migration assessment, colony-forming capacity and gene expression analysis. DNR combination with DNC resulted in increased apoptosis to necrosis ratio compared to DNR alone. In addition, the migration and colony-forming capacity were at the minimal range when DNC was combined with DNR. Combined treatment decreased the expression level of MDR-1, hTERT and Bcl-2 genes significantly. In addition, the ratio of Bax/Bcl2 gene expression significantly increased. Our analysis by free curcumin, dendrosomes and DNC also showed that dendrosomes do not have any significant cytotoxic effect on the A549 cells, suggesting that this carrier has a high potential for enhancing the curcumin's biological effects. Conclusion Our observations suggest that the DNC formulation of curcumin synergistically enhances the antineoplastic effect of DNR on the A549 cell line through the modulation of apoptosis/necrosis ratio, as well as Bax/Bcl2 ratio, MDR-1 and hTERT gene expression.
Collapse
Affiliation(s)
- Seyed Sadegh Eslami
- Student Research Committee, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
- Department of Medical Biotechnology, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Davod Jafari
- Student Research Committee, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
- Department of Medical Biotechnology, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Abbas Ghotaslou
- Department of Clinical Laboratory Sciences, School of Allied Medical Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Moein Amoupour
- Department of Medical Biotechnology, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Amir Asri Kojabad
- Department of Clinical Laboratory Sciences, School of Allied Medical Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Rasool Jafari
- Department of Medical Parasitology and Mycology, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Navid Mousazadeh
- Department of Medical Biotechnology, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Parastoo Tarighi
- Department of Medical Biotechnology, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Majid Sadeghizadeh
- Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
8
|
Tseng TY, Lee CH, Lee HL, Su CY, Kao CY, Tsai JP, Hsieh YH. Licochalcone A Suppresses Renal Cancer Cell Proliferation and Metastasis by Engagement of Sp1-Mediated LC3 Expression. Pharmaceutics 2023; 15:pharmaceutics15020684. [PMID: 36840005 PMCID: PMC9966374 DOI: 10.3390/pharmaceutics15020684] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 02/14/2023] [Accepted: 02/15/2023] [Indexed: 02/19/2023] Open
Abstract
Licochalcone A (LicA) is a strong anti-inflammatory, antioxidant, and anticarcinogenic substance that is useful against a variety of human malignancies. However, its precise mechanism in mediating the development of renal cell carcinoma (RCC) is not entirely understood. In this work, LicA was discovered to limit cell growth and survival, induce cell cycle arrest, promote autophagy and LC3B expression, and inhibit the migration and invasion of RCC cells. In addition, the proliferation, migration, and invasion inhibited by LicA were restored by the transfection of siRNA-LC3. The effects of LC3B on the metastatic phenotype of ACHN cells was enhanced with the overexpression of Sp1 or suppressed by inhibiting the phosphorylation of FAK and Src. Finally, LicA showed antitumor properties against RCC in an in vivo xenograft model. In conclusion, our study demonstrated the chemotherapeutic potential of LicA on proliferation, migration, invasion, and autophagy through the activation of LC3B expression, ultimately modulating FAK/Src signaling pathway-mediated Sp1 expression. These findings illustrate the novel role and molecular mechanisms of LicA against RCC cells.
Collapse
Affiliation(s)
- Tsai-Yi Tseng
- Institute of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan
- Division of Pediatric Surgery, Department of Surgery, Children’s Hospital of China Medical University, Taichung 404333, Taiwan
| | - Chien-Hsing Lee
- Division of Pediatric Surgery, Department of Surgery, Children’s Hospital of China Medical University, Taichung 404333, Taiwan
- School of Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung 404333, Taiwan
| | - Hsiang-Lin Lee
- Department of Surgery, Chung Shan Medical University Hospital, Taichung 40201, Taiwan
- School of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan
| | - Chien-Yu Su
- Institute of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan
| | - Cheng-Yen Kao
- Institute of Microbiology and Immunology, College of Life Sciences, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan
| | - Jen-Pi Tsai
- School of Medicine, Tzu Chi University, Hualien 970374, Taiwan
- Division of Nephrology, Department of Internal Medicine, Dalin Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Chiayi 62247, Taiwan
- Correspondence: (J.-P.T.); (Y.-H.H.)
| | - Yi-Hsien Hsieh
- Institute of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung 40201, Taiwan
- Correspondence: (J.-P.T.); (Y.-H.H.)
| |
Collapse
|
9
|
Dhamodharan J, Sekhar G, Muthuraman A. Epidermal Growth Factor Receptor Kinase Inhibitor Ameliorates β-Amyloid Oligomer-Induced Alzheimer Disease in Swiss Albino Mice. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27165182. [PMID: 36014421 PMCID: PMC9412386 DOI: 10.3390/molecules27165182] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Revised: 08/04/2022] [Accepted: 08/11/2022] [Indexed: 11/16/2022]
Abstract
Alzheimer's disease (AD) is one of the major neurodegenerative disorders, and its incidence increases globally every year. Currently, available AD drugs symptomatically treat AD with multiple adverse effects. Gefitinib (GE) is an epidermal growth factor receptor (EGFR) kinase inhibitor. EGFR is the preferred target for the treatment of AD, whereas the effect of GE in AD conditions is limited. The present study was designed to explore the ameliorative potential of GE in Aβ1-42 oligomer-induced neurotoxicity in AD mice. AD was induced by intracerebroventricular (i.c.v.) injection of Aβ1-42 oligomer (4 μg/4 μL) into the lateral ventricles of the mouse brain. The test compound, i.e., GE (2 and 4 mg/kg of body weight), was administered orally on days 10, 13, 16, 19, 22, 25, and 28, and the reference drug, i.e., donepezil (DP, 2 mg/kg), was administered orally from the 10th to 28th days. The behavioral changes were screened by the Morris water maze (MWM) test. Furthermore, biomarkers i.e., brain acetylcholinesterase (AChE), thiobarbituric acid reactive substances (TBARS), and reduced glutathione (GSH) levels were estimated from brain samples. The AD-associated histopathological changes were analyzed by hematoxylin and eosin staining. The administration of GE significantly ameliorated the AD-associated behavioral, biochemical, and histopathological changes. The ameliorative effect of GE against the Aβ1-42 oligomer-associated neurotoxicity was due to its potent inhibition of EGFR kinase activation, as well as its antioxidant and antilipid peroxidative effect.
Collapse
Affiliation(s)
- Jagadeesh Dhamodharan
- Unit of Anatomy, Faculty of Medicine, AIMST University, Semeling, Bedong 08100, Kedah, Malaysia
- Department of Pathology, Faculty of Medicine, Saveetha Institute of Medical & Technical Sciences (SIMATS), Saveetha University, Chennai 602105, Tamilnadu, India
| | - Ganthimathy Sekhar
- Department of Pathology, Faculty of Medicine, Saveetha Institute of Medical & Technical Sciences (SIMATS), Saveetha University, Chennai 602105, Tamilnadu, India
| | - Arunachalam Muthuraman
- Unit of Pharmacology, Faculty of Pharmacy, AIMST University, Bedong 08100, Kedah, Malaysia
- Correspondence:
| |
Collapse
|
10
|
Xie H, Jing R, Liao X, Chen H, Xie X, Dai H, Pan L. Arecoline promotes proliferation and migration of human HepG2 cells through activation of the PI3K/AKT/mTOR pathway. Hereditas 2022; 159:29. [PMID: 35836300 PMCID: PMC9281068 DOI: 10.1186/s41065-022-00241-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 06/23/2022] [Indexed: 11/26/2022] Open
Abstract
Background Arecoline is a well-known risk factor for oral submucosal fibrosis and cancer. However, the mechanistic correlation between arecoline and hepatocellular cancer remains elusive. Here, we investigated the effect of arecoline on the proliferation and migration of human HepG2 hepatoma cells and its potential oncogenic mechanisms. Methods Bioinformatic technologies were used to identify the deferentially expressed miRNAs (DE-miRNAs) and hub target genes of arecoline-induced cancers. These DE-miRNAs, hub genes and pathway were proved in arecoline-treated HepG2 cells. Results A total of 86 DE-miRNAs and 460 target genes were identified. These target genes are associated with DNA-templated regulation of transcription and other biological processes. Significant molecular functions were protein binding, calcium ion binding, and enrichment in the nucleus and cytoplasm. These genes are involved in the PI3K-AKT pathway. CDK1, CCND1, RAF1, CDKN1B and BTRC were defined as the top 5 hub target genes, and patients with high expression of CDK1 showed poor prognosis. Compared with control group, 2.5 µM arecoline treatment increased the proliferation and migration ability of the HepG2 cells. Treatment with 2.5 µM arecoline increased the levels of miR-21-3p, miR-21-5p and miR-1267, upregulated the expression of PI3K-AKT pathway factors, CDK1, CCND1 but decreased RAF1 expression. Conclusion A low concentration arecoline can induce the proliferation and migration of HepG2 cells, with the potential mechanism of action linked to high levels of exosomal miR-21 and miR-1267, activation of the PI3K-AKT pathway, upregulation of CDK1 and CCND1, and downregulation of RAF1.
Collapse
Affiliation(s)
- Hai Xie
- Department of Anesthesiology, Guangxi Medical University Affiliated Cancer Hospital, He Di Rd No.71, Nanning, 530021, PR China.,Guangxi Key Laboratory for Basic Science and Prevention of Perioperative Organ Disfunction, Guangxi Medical University Cancer Hospital, Nanning, China.,Department of Anesthesiology, The First Affiliated Hospital of Hainan Medical College, Haikou, China
| | - Ren Jing
- Department of Anesthesiology, Guangxi Medical University Affiliated Cancer Hospital, He Di Rd No.71, Nanning, 530021, PR China.,Guangxi Key Laboratory for Basic Science and Prevention of Perioperative Organ Disfunction, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Xiaoting Liao
- Department of Anesthesiology, Guangxi Medical University Affiliated Cancer Hospital, He Di Rd No.71, Nanning, 530021, PR China.,Guangxi Key Laboratory for Basic Science and Prevention of Perioperative Organ Disfunction, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Haishao Chen
- Department of Anesthesiology, Guangxi Medical University Affiliated Cancer Hospital, He Di Rd No.71, Nanning, 530021, PR China.,Guangxi Key Laboratory for Basic Science and Prevention of Perioperative Organ Disfunction, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Xianlong Xie
- Department of Anesthesiology, Guangxi Medical University Affiliated Cancer Hospital, He Di Rd No.71, Nanning, 530021, PR China.,Guangxi Key Laboratory for Basic Science and Prevention of Perioperative Organ Disfunction, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Huijun Dai
- Department of Anesthesiology, Guangxi Medical University Affiliated Cancer Hospital, He Di Rd No.71, Nanning, 530021, PR China.,Guangxi Key Laboratory for Basic Science and Prevention of Perioperative Organ Disfunction, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Linghui Pan
- Department of Anesthesiology, Guangxi Medical University Affiliated Cancer Hospital, He Di Rd No.71, Nanning, 530021, PR China. .,Guangxi Key Laboratory for Basic Science and Prevention of Perioperative Organ Disfunction, Guangxi Medical University Cancer Hospital, Nanning, China.
| |
Collapse
|
11
|
Calaf GM, Crispin LA, Muñoz JP, Aguayo F, Bleak TC. Muscarinic Receptors Associated with Cancer. Cancers (Basel) 2022; 14:cancers14092322. [PMID: 35565451 PMCID: PMC9100020 DOI: 10.3390/cancers14092322] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 04/26/2022] [Accepted: 04/30/2022] [Indexed: 02/01/2023] Open
Abstract
Simple Summary Recently, cancer research has described the presence of the cholinergic machinery, specifically muscarinic receptors, in a wide variety of cancers due to their activation and signaling pathways associated with tumor progression and metastasis, providing a wide overview of their contribution to different cancer formation and development for new antitumor targets. This review focused on determining the molecular signatures associated with muscarinic receptors in breast and other cancers and the need for pharmacological, molecular, biochemical, technological, and clinical approaches to improve new therapeutic targets. Abstract Cancer has been considered the pathology of the century and factors such as the environment may play an important etiological role. The ability of muscarinic agonists to stimulate growth and muscarinic receptor antagonists to inhibit tumor growth has been demonstrated for breast, melanoma, lung, gastric, colon, pancreatic, ovarian, prostate, and brain cancer. This work aimed to study the correlation between epidermal growth factor receptors and cholinergic muscarinic receptors, the survival differences adjusted by the stage clinical factor, and the association between gene expression and immune infiltration level in breast, lung, stomach, colon, liver, prostate, and glioblastoma human cancers. Thus, targeting cholinergic muscarinic receptors appears to be an attractive therapeutic alternative due to the complex signaling pathways involved.
Collapse
Affiliation(s)
- Gloria M. Calaf
- Instituto de Alta Investigación, Universidad de Tarapacá, Arica 1000000, Chile; (L.A.C.); (J.P.M.); (T.C.B.)
- Correspondence:
| | - Leodan A. Crispin
- Instituto de Alta Investigación, Universidad de Tarapacá, Arica 1000000, Chile; (L.A.C.); (J.P.M.); (T.C.B.)
| | - Juan P. Muñoz
- Instituto de Alta Investigación, Universidad de Tarapacá, Arica 1000000, Chile; (L.A.C.); (J.P.M.); (T.C.B.)
| | - Francisco Aguayo
- Laboratorio de Oncovirología, Programa de Virología, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago 8380000, Chile;
| | - Tammy C. Bleak
- Instituto de Alta Investigación, Universidad de Tarapacá, Arica 1000000, Chile; (L.A.C.); (J.P.M.); (T.C.B.)
| |
Collapse
|
12
|
Chu M, Fu G, Deng J, Wang R, Fan Q, Chen Z, Lu J, Liu XA. Evaluation of the inhalation toxicity of arecoline benzoate aerosol in rats. J Appl Toxicol 2022; 42:1396-1410. [PMID: 35170056 DOI: 10.1002/jat.4303] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 02/05/2022] [Accepted: 02/09/2022] [Indexed: 11/10/2022]
Abstract
Arecoline is a pharmacologically active alkaloid isolated from Areca catechu. There are no published data available regarding the inhalation toxicity of arecoline in animals. This study aimed to evaluate the inhalation toxicity of arecoline in vitro and in vivo. For this purpose, arecoline benzoate (ABA) salt was prepared to stabilize arecoline in an aerosol. The MTT assay determined the half-maximal inhibitory concentration values of ABA and arecoline in A549 cell proliferation to be 832 μg/ml and 412 μg/ml, respectively. The toxicity of acute and subacute inhalation in Sprague-Dawley rats was evaluated using the guidelines of the Organization for Economic Cooperation and Development. For acute inhalation, the median lethal concentration value of ABA solvent was >5175 mg/m3 . After the exposure and during the recovery period, no treatment-related clinical signs were observed. In the 28-Day inhalation toxicity test, daily nose-only exposure to 2510 mg/m3 aerosol of the ABA solvent contained 75 mg/m3 ABA for male rats and 375 mg/m3 ABA for female rats, which caused no observed adverse effects, except for the decreased body weight gain in male rats exposed to 375 mg/m3 ABA. In this study, the no observed adverse effect level (NOAEL) for the 28-Day repeated dose inhalation of ABA aerosol was calculated to be around 13 mg/kg/day for male rats and 68.8 mg/kg/day for female rats, respectively.
Collapse
Affiliation(s)
- Ming Chu
- Laboratory of Life and Health Sciences, Shenzhen First Union Technology Co., Ltd, Shenzhen, China.,Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, CAS Key Laboratory of Brain Connectome and Manipulation, Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, China
| | - Guofeng Fu
- Laboratory of Life and Health Sciences, Shenzhen First Union Technology Co., Ltd, Shenzhen, China.,Laboratory of Life Sciences, Shenzhen Icybetel Biotechnology Co, Ltd, Shenzhen, China
| | - Jingjing Deng
- Laboratory of Life and Health Sciences, Shenzhen First Union Technology Co., Ltd, Shenzhen, China
| | - Ruoxi Wang
- Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, CAS Key Laboratory of Brain Connectome and Manipulation, Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, China
| | - Qiming Fan
- GuangdongZhongkeEnHealth Science and Technology Co., Ltd., Foshan, China
| | - Zuxin Chen
- Shenzhen Key Laboratory of Drug Addiction, Shenzhen Neher Neural Plasticity Laboratory, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences (CAS); Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, 518055, China; University of Chinese Academy of Sciences, Beijing, China
| | - Jin Lu
- Laboratory of Life and Health Sciences, Shenzhen First Union Technology Co., Ltd, Shenzhen, China.,Laboratory of Life Sciences, Shenzhen Icybetel Biotechnology Co, Ltd, Shenzhen, China
| | - Xin-An Liu
- Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, CAS Key Laboratory of Brain Connectome and Manipulation, Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, 518055, China; University of the Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
13
|
Chen CY, Lin H, Cheng PT, Cheng YC, Oner M, Li YH, Chen MC, Wu JH, Chang TC, Celik A, Liu FL, Wang HY, Lai CH, Hsieh JT. Antrodia salmonea extract inhibits cell proliferation through regulating cell cycle arrest and apoptosis in prostate cancer cell lines. CHINESE J PHYSIOL 2022; 65:209-214. [DOI: 10.4103/cjp.cjp_78_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
14
|
Yue CH, Oner M, Chiu CY, Chen MC, Teng CL, Wang HY, Hsieh JT, Lai CH, Lin H. RET Regulates Human Medullary Thyroid Cancer Cell Proliferation through CDK5 and STAT3 Activation. Biomolecules 2021; 11:biom11060860. [PMID: 34207842 PMCID: PMC8229599 DOI: 10.3390/biom11060860] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 06/04/2021] [Accepted: 06/07/2021] [Indexed: 12/11/2022] Open
Abstract
Medullary thyroid cancer (MTC) is a neuroendocrine tumor that arises from the parafollicular C-cells, which produces the hormone calcitonin. RET is a transmembrane receptor protein-tyrosine kinase, which is highly expressed in MTC. Our previous studies reported that cyclin-dependent kinase 5 (CDK5) plays a crucial role in cancer progression, including MTC. However, the role of CDK5 in GDNF-induced RET signaling in medullary thyroid cancer proliferation remains unknown. Here, we investigated RET activation and its biochemically interaction with CDK5 in GDNF-induced medullary thyroid cancer proliferation. Our results demonstrated that GDNF stimulated RET phosphorylation and thus subsequently resulted in CDK5 activation by its phosphorylation. Activated CDK5 further caused STAT3 activation by its specific phosphorylation at Ser727. Moreover, we also found that GDNF treatment enhanced ERK1/2 and EGR1 activity, which is involved in p35 activation. Interestingly, we identified for the first time that CDK5 physically interacted with RET protein in MTC. Overall, our results provide a new mechanism for medullary thyroid cancer cell proliferation, suggesting that targeting CDK5 may be a promising therapeutic candidate for human medullary thyroid cancer in the near future.
Collapse
Affiliation(s)
- Chia-Herng Yue
- Department of Surgery, Tung’s Taichung Metro Harbor Hospital, Taichung 435403, Taiwan;
| | - Muhammet Oner
- Department of Life Sciences, National Chung Hsing University, Taichung 402204, Taiwan; (M.O.); (C.-Y.C.)
| | - Chih-Yuan Chiu
- Department of Life Sciences, National Chung Hsing University, Taichung 402204, Taiwan; (M.O.); (C.-Y.C.)
| | - Mei-Chih Chen
- Translational Cell Therapy Center, Department of Medical Research, China Medical University Hospital, Taichung 404332, Taiwan;
| | - Chieh-Lin Teng
- Division of Hematology/Medical Oncology, Department of Medicine, Taichung Veterans General Hospital, Taichung 40201, Taiwan;
- Department of Life Science, Tunghai University, Taichung 40704, Taiwan
- School of Medicine, Chung Shan Medical University, Taichung 402, Taiwan
| | - Hsin-Yi Wang
- Department of Nuclear Medicine, Taichung Veterans General Hospital, Taichung 40705, Taiwan;
| | - Jer-Tsong Hsieh
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA;
| | - Chih-Ho Lai
- Department of Microbiology and Immunology, Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan;
| | - Ho Lin
- Department of Life Sciences, National Chung Hsing University, Taichung 402204, Taiwan; (M.O.); (C.-Y.C.)
- Ph.D. Program in Translational Medicine, National Chung Hsing University, Taichung 402204, Taiwan
- Rong Hsing Research Center for Translational Medicine, National Chung Hsing University, Taichung 402204, Taiwan
- Correspondence: ; Tel.: +886-4-22840-416 (ext. 311); Fax: +886-4-22874-740
| |
Collapse
|
15
|
Albuquerque C, Manguinhas R, Costa JG, Gil N, Codony-Servat J, Castro M, Miranda JP, Fernandes AS, Rosell R, Oliveira NG. A narrative review of the migration and invasion features of non-small cell lung cancer cells upon xenobiotic exposure: insights from in vitro studies. Transl Lung Cancer Res 2021; 10:2698-2714. [PMID: 34295671 PMCID: PMC8264350 DOI: 10.21037/tlcr-21-121] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Accepted: 05/10/2021] [Indexed: 01/03/2023]
Abstract
Lung cancer (LC) is the leading cause of cancer deaths worldwide, being non-small lung cancer (NSCLC) sub-types the most prevalent. Since most LC cases are only detected during the last stage of the disease the high mortality rate is strongly associated with metastases. For this reason, the migratory and invasive capacity of these cancer cells as well as the mechanisms involved have long been studied to uncover novel strategies to prevent metastases and improve the patients’ prognosis. This narrative review provides an overview of the main in vitro migration and invasion assays employed in NSCLC research. While several methods have been developed, experiments using conventional cell culture models prevailed, specifically the wound-healing and the transwell migration and invasion assays. Moreover, it is provided herewith a summary of the available information concerning chemical contaminants that may promote the migratory/invasive properties of NSCLC cells in vitro, shedding some light on possible LC risk factors. Most of the reported agents with pro-migration/invasion effects derive from cigarette smoking [e.g., Benzo(a)pyrene and cadmium] and air pollution. This review further presents several studies in which different dietary/plant-derived compounds demonstrated to impair migration/invasion processes in NSCLC cells in vitro. These chemicals that have been proposed as anti-migratory consisted mainly of natural bioactive substances, including polyphenols non-flavonoids, flavonoids, bibenzyls, terpenes, alkaloids, and steroids. Some of these compounds may eventually represent novel therapeutic strategies to be considered in the future to prevent metastasis formation in LC, which highlights the need for additional in vitro methodologies that more closely resemble the in vivo tumor microenvironment and cancer cell interactions. These studies along with adequate in vivo models should be further explored as proof of concept for the most promising compounds.
Collapse
Affiliation(s)
- Catarina Albuquerque
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisboa, Portugal
| | - Rita Manguinhas
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisboa, Portugal
| | - João G Costa
- CBIOS, Universidade Lusófona's Research Center for Biosciences & Health Technologies, Lisboa, Portugal
| | - Nuno Gil
- Lung Cancer Unit, Champalimaud Centre for the Unknown, Lisboa, Portugal
| | - Jordi Codony-Servat
- Laboratory of Oncology/Pangaea Oncology S.L., Quirón-Dexeus University Institute, Barcelona, Spain
| | - Matilde Castro
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisboa, Portugal
| | - Joana P Miranda
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisboa, Portugal
| | - Ana S Fernandes
- CBIOS, Universidade Lusófona's Research Center for Biosciences & Health Technologies, Lisboa, Portugal
| | - Rafael Rosell
- Laboratory of Oncology/Pangaea Oncology S.L., Quirón-Dexeus University Institute, Barcelona, Spain.,Laboratory of Cellular and Molecular Biology, Institute for Health Science Research Germans Trias i Pujol (IGTP), Campus Can Ruti, Barcelona, Spain.,Internal Medicine Department, Universitat Autónoma de Barcelona, Campus de la UAB, Barcelona, Spain
| | - Nuno G Oliveira
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisboa, Portugal
| |
Collapse
|
16
|
Identification of novel biomarkers and candidate small molecule drugs in rheumatoid arthritis and osteoarthritis based on bioinformatics analysis of high-throughput data. Biosci Rep 2021; 40:226645. [PMID: 33325525 PMCID: PMC7744737 DOI: 10.1042/bsr20193823] [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: 11/24/2019] [Revised: 09/14/2020] [Accepted: 10/07/2020] [Indexed: 02/07/2023] Open
Abstract
Background: Rheumatoid arthritis (RA) and osteoarthritis (OA) are two major types of joint diseases. The present study aimed to identify hub genes involved in the pathogenesis and further explore the potential treatment targets of RA and OA. Methods: The gene expression profile of GSE12021 was downloaded from Gene Expression Omnibus (GEO). Total 31 samples (12 RA, 10 OA and 9 NC samples) were used. The differentially expressed genes (DEGs) in RA versus NC, OA versus NC and RA versus OA groups were screened using limma package. We also verified the DEGs in GSE55235 and GSE100786. Functional annotation and protein–protein interaction (PPI) network construction of OA‐ and RA‐specific DEGs were performed. Finally, the candidate small molecules as potential drugs to treat RA and OA were predicted in CMap database. Results: 165 up-regulated and 163 down-regulated DEGs between RA and NC samples, 73 up-regulated and 293 down-regulated DEGs between OA and NC samples, 92 up-regulated and 98 down-regulated DEGs between RA and OA samples were identified. Immune response and TNF signaling pathway were significantly enriched pathways for RA‐ and OA‐specific DEGs, respectively. The hub genes were mainly associated with ‘Primary immunodeficiency’ (RA vs. NC group), ‘Ribosome’ (OA vs. NC group), and ‘Chemokine signaling pathway’ (RA vs. OA group). Arecoline and Cefamandole were the most promising small molecule to reverse the RA and OA gene expression. Conclusion: Our findings suggest new insights into the underlying pathogenesis of RA and OA, which may improve the diagnosis and treatment of these intractable chronic diseases.
Collapse
|
17
|
Zhu YG, Lv YX, Guo CY, Xiao ZM, Jiang QG, Kuang H, Zhang WH, Hu P. Harmine inhibits the proliferation and migration of glioblastoma cells via the FAK/AKT pathway. Life Sci 2021; 270:119112. [DOI: 10.1016/j.lfs.2021.119112] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 01/09/2021] [Accepted: 01/19/2021] [Indexed: 01/01/2023]
|
18
|
Chen Q, Jiao J, Wang Y, Mai Z, Ren J, He S, Li X, Chen Z. Egr-1 mediates low-dose arecoline induced human oral mucosa fibroblast proliferation via transactivation of Wnt5a expression. BMC Mol Cell Biol 2020; 21:80. [PMID: 33167868 PMCID: PMC7653895 DOI: 10.1186/s12860-020-00325-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Accepted: 10/27/2020] [Indexed: 01/08/2023] Open
Abstract
Background Arecoline is an alkaloid natural product found in the areca nut that can induce oral submucous fibrosis and subsequent development of cancer. However, numerous studies have shown that arecoline may inhibit fibroblast proliferation and prevent collagen synthesis. Results High doses of arecoline (> 32 μg/ml) could inhibit human oral fibroblast proliferation, while low doses of arecoline (< 16 μg/ml) could promote the proliferation of human oral fibroblasts. Wnt5a was found to be both sufficient and necessary for the promotion of fibroblast proliferation. Egr-1 could mediate the expression of Wnt5a in fibroblasts, while NF-κB, FOXO1, Smad2, and Smad3 did not. Treatment with siRNAs specific to Egr-1, Egr inhibitors, or Wnt5a antibody treatment could all inhibit arecoline-induced Wnt5a upregulation and fibroblast proliferation. Conclusions Egr-1 mediates the effect of low dose arecoline treatment on human oral mucosa fibroblast proliferation by transactivating the expression of Wnt5a. Therefore, Egr inhibitors and Wnt5a antibodies are potential therapies for treatment of oral submucosal fibrosis and oral cancer.
Collapse
Affiliation(s)
- Qiang Chen
- Department of Stomatology, the Third Affiliated Hospital of Sun Yat-sen University, No.600 Tianhe road, Guangzhou, 510630, China
| | - Jiuyang Jiao
- Department of Oral & Maxillofacial Surgery & Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Youyuan Wang
- Department of Oral & Maxillofacial Surgery & Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zhihui Mai
- Department of Stomatology, the Third Affiliated Hospital of Sun Yat-sen University, No.600 Tianhe road, Guangzhou, 510630, China
| | - Jing Ren
- Department of Stomatology, the Third Affiliated Hospital of Sun Yat-sen University, No.600 Tianhe road, Guangzhou, 510630, China
| | - Sijie He
- The fourth people's hospital of Nanhai district of Foshan city, Foshan, China.
| | - Xiaolan Li
- Guanghua School of stomatology & hospital of stomatology, Guangdong province key laboratory of stomatology, Sun Yat-sen University, Guangzhou, China.
| | - Zheng Chen
- Department of Stomatology, the Third Affiliated Hospital of Sun Yat-sen University, No.600 Tianhe road, Guangzhou, 510630, China.
| |
Collapse
|
19
|
Kim D, Illeperuma RP, Kim J. The Protective Effect of Antioxidants in Areca Nut Extract-Induced Oral Carcinogenesis. Asian Pac J Cancer Prev 2020; 21:2447-2452. [PMID: 32856877 PMCID: PMC7771929 DOI: 10.31557/apjcp.2020.21.8.2447] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Indexed: 12/14/2022] Open
Abstract
Objective: Oral submucous fibrosis (OSF) is the premalignant disorder associated with fibrosis and epithelial atrophy. Areca Nut (AN) is the most significant risk factors for OSF. However, the molecular mechanism behind AN induced OSF remains unclear, and there exists no effective treatment for the malignant disorder. We aimed to investigate whether AN-extract causes epithelial-mesenchymal transition (EMT) in oral keratinocytes, and evaluated the therapeutic potential of antioxidants. Methods: The HPV16 E6/E7-transfected immortalized human oral keratinocytes (IHOK) were employed in the present study. For the preparation of AN-extract, dried AN was dissolved in distilled water overnight. The solution was centrifuged and the supernatant was collected for further use. For the determination of change in cytokine levels, ELISA was performed. To investigate EMT-related protein expression and phenotype, immunoblot and immunofluorescence were performed. Results: Among tumor-promoting cytokines (Gro-α, IL-6 and IL-8), IL-6 was remarkably increased by AN in IHOK. AN-extract induced EMT phenotypes, such as cell elongation, up-regulation of vimentin and snail. After treatment with neutralizing antibody of IL-6, AN-induced snail expression was reduced remarkably. Collectively, AN-extract induced IL-6 expression and mediated EMT. The use of antioxidants (EGCG, glutathione and NAC) significantly reduced IL-6 expression in AN-treated IHOK. Also, AN-decreased E-cadherin and increased vimentin were reversed by antioxidants, indicating that the effectiveness of antioxidants in inhibiting IL-6-induced EMT by AN. Conclusion: AN promotes EMT and antioxidants interrupt AN-induced-EMT in oral keratinocytes. Consequently, it is proposed that antioxidants could prevent AN-induced carcinogenesis and function as a prototype for developing therapeutic interventions of OSF.
Collapse
Affiliation(s)
- Dokyeong Kim
- Department of Dental hygiene, Jeonju Kijeon College, Jeonju, Republic of Korea
| | - Rasika Pawiththra Illeperuma
- Department of Medical Laboratory Science, Faculty of Allied Health Sciences, University of Peradeniya, Peradeniya, Sri Lanka
| | - Jin Kim
- Department of Oral pathology, Oral Cancer Research Institute, Yonsei University College of Dentistry, Seoul, Republic of Korea
| |
Collapse
|
20
|
Adamski Z, Blythe LL, Milella L, Bufo SA. Biological Activities of Alkaloids: From Toxicology to Pharmacology. Toxins (Basel) 2020; 12:toxins12040210. [PMID: 32224853 PMCID: PMC7232379 DOI: 10.3390/toxins12040210] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 03/25/2020] [Indexed: 12/20/2022] Open
Affiliation(s)
- Zbigniew Adamski
- Department of Animal Physiology and Development/Electron and Confocal Microscope Laboratory, Faculty of Biology, Adam Mickiewicz University, 61-614 Poznań, Poland
- Correspondence: (Z.A.); (S.A.B.)
| | - Linda L. Blythe
- Department of Veterinary Medicine, Oregon State University, Corvallis, 97331 OR, USA;
| | - Luigi Milella
- Department of Science, University of Basilicata, 85100 Potenza, Italy;
| | - Sabino A. Bufo
- Department of Science, University of Basilicata, 85100 Potenza, Italy;
- Correspondence: (Z.A.); (S.A.B.)
| |
Collapse
|
21
|
Huang K, Gao N, Bian D, Zhai Q, Yang P, Li M, Wang X. Correlation between FAK and EGF-Induced EMT in Colorectal Cancer Cells. JOURNAL OF ONCOLOGY 2020; 2020:5428920. [PMID: 32148496 PMCID: PMC7048944 DOI: 10.1155/2020/5428920] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/02/2019] [Revised: 01/14/2020] [Accepted: 01/23/2020] [Indexed: 12/31/2022]
Abstract
Epithelial-mesenchymal transition (EMT) plays an important role in the invasion and metastasis of colorectal cancer, which is mediated by FAK and EGF. However, whether FAK participates in EMT in colorectal cancer cells through the EGF/EGFR signaling pathway remains unknown. The aim of this study was to investigate the effector mechanisms of FAK in the process of EGF-induced EMT in colorectal cancer cells and to determine whether miR-217 is involved in this process. Caco-2 cancer cells were routinely cultured with and without treatment with 100 ng/mL EGF, and changes in cell morphology were observed using an inverted microscope. In addition, a transwell assay was used to detect cell migration under the condition of EGF treatment. The expression of FAK, pFAK, E-cadherin, vimentin, and β actin was assessed by western blotting, and the expression of miR-217 was assessed using real-time PCR. We found that EGF induced EMT in colorectal cancer cells and enhanced cell migration and invasion ability. Moreover, FAK was involved in the EGF-induced EMT of colorectal cancer cells. EGF upregulated the expression of E-cadherin in colorectal cancer cells by activating FAK, and miR-217 was found to participate in EGF-induced EMT in colorectal cancer cells. Our findings indicate that EGF induces EMT in colorectal cancer cells by activating FAK, and miR-217 is involved in the EGF/FAK/E-cadherin signaling pathway.
Collapse
Affiliation(s)
- Kun Huang
- Department of Ultrasonic Diagnosis, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Ningning Gao
- Department of Ultrasonic Diagnosis, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Donglin Bian
- Department of Ultrasonic Diagnosis, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Qixi Zhai
- Department of Ultrasonic Diagnosis, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Puxu Yang
- Department of Ultrasonic Diagnosis, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Mingwei Li
- Department of Ultrasonic Diagnosis, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Xuemei Wang
- Department of Ultrasonic Diagnosis, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China
| |
Collapse
|
22
|
Gupta AK, Tulsyan S, Thakur N, Sharma V, Sinha DN, Mehrotra R. Chemistry, metabolism and pharmacology of carcinogenic alkaloids present in areca nut and factors affecting their concentration. Regul Toxicol Pharmacol 2020; 110:104548. [PMID: 31805361 DOI: 10.1016/j.yrtph.2019.104548] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Revised: 11/27/2019] [Accepted: 12/01/2019] [Indexed: 12/28/2022]
Abstract
Areca Nut (AN), the seed of tropical palm tree Areca catechu, is a widely chewed natural product with estimated 600 million users across the world. Various AN products, thriving in the market, portray 'Areca nut' or 'Supari' as mouth freshener and safe alternative to smokeless tobacco. Unfortunately, AN is identified as a Group 1 human carcinogen by International Agency for Research on Cancer (IARC). Wide variation in the level of alkaloids, broadly ranging from 2 to 10 mg/gm dry weight, is observed in diverse variety of AN sold worldwide. For the first time, various factors influencing the formation of carcinogenic alkaloids in AN at various stages, including during the growth, processing, and storage of the nut, are discussed. Current review illustrates the mechanism of cancer induction by areca alkaloids in humans and also compiles dose-dependent pharmacology and toxicology data of arecoline, the most potent carcinogenic alkaloid in AN. Careful monitoring of the arecoline content in AN can potentially be used as a tool in product surveillance studies to identify the variations in characteristics of various AN sample sold worldwide. The article will help to generate public awareness and sensitize the government bodies to initiate campaigns against AN use and addiction.
Collapse
Affiliation(s)
- Alpana K Gupta
- Division of Preventive Oncology, ICMR-National Institute of Cancer Prevention and Research, Department of Health Research (Govt. of India), I-7, Sector-39, Noida, G.B. Nagar, 201301, U.P, India
| | - Sonam Tulsyan
- Division of Preventive Oncology, ICMR-National Institute of Cancer Prevention and Research, Department of Health Research (Govt. of India), I-7, Sector-39, Noida, G.B. Nagar, 201301, U.P, India
| | - Nisha Thakur
- Division of Molecular Diagnostics, ICMR-National Institute of Cancer Prevention and Research, Department of Health Research (Govt. of India), I-7, Sector-39, Noida, G.B. Nagar, 201301, U.P, India
| | - Vishwas Sharma
- Division of Preventive Oncology, ICMR-National Institute of Cancer Prevention and Research, Department of Health Research (Govt. of India), I-7, Sector-39, Noida, G.B. Nagar, 201301, U.P, India
| | | | - Ravi Mehrotra
- ICMR- India Cancer Research Consortium, Department of Health Research (Govt. of India), 1- Red Cross Road, New Delhi, 110001, India.
| |
Collapse
|
23
|
Synthesis and screening of novel anthraquinone−quinazoline multitarget hybrids as promising anticancer candidates. Future Med Chem 2020; 12:111-126. [PMID: 31718309 DOI: 10.4155/fmc-2019-0230] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Aim: The EGF receptor (EGFR) is overexpressed in multiple epithelial-derived cancers and is considered to be a vital target closely associated with cancer therapy. In this study, a series of novel anthraquinone−quinazoline hybrids targeting several vital sites for cancer therapy were designed and synthesized. Methodology & results: Most of the synthesized hybrids demonstrated excellent antiproliferative activity and downregulation of the expression of EGFR. The most promising compound 7d showed the strongest antiproliferation activity; this compound significantly downregulated the expression of p-EGFR protein, induced a remarkable apoptosis effect, promoted the rearrangement of F-actin filaments and destruction of cytoskeleton, induced DNA damage and enhanced radiosensitivity of A549 cells. Conclusion: The novel anthraquinone−quinazoline hybrid 7d emerges as an anticancer drug candidate with promising multitargeted biological activities.
Collapse
|
24
|
Prince GMSH, Yang TY, Lin H, Chen MC. Mechanistic insight of cyclin-dependent kinase 5 in modulating lung cancer growth. CHINESE J PHYSIOL 2019; 62:231-240. [PMID: 31793458 DOI: 10.4103/cjp.cjp_67_19] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Lung harbors the growth of primary and secondary tumors. Even though numerous factors regulate the complex signal transduction and cytoskeletal remodeling toward the progression of lung cancer, cyclin-dependent kinase 5 (Cdk5), a previously known kinase in the central nervous system, has raised much attention in the recent years. Patients with aberrant Cdk5 expression also lead to poor survival. Cdk5 has already been employed in various cellular processes which shape the fate of cancer. In lung cancer, Cdk5 mainly regulates tumor suppressor genes, carcinogenesis, cytoskeletal remodeling, and immune checkpoints. Inhibiting Cdk5 by using drugs, siRNA or CRISP-Cas9 system has rendered crucial therapeutic advantage in the combat against lung cancer. Thus, the relation of Cdk5 to lung cancer needs to be addressed in detail. In this review, we will discuss various cellular events modulated by Cdk5 and we will go further into their underlying mechanism in lung cancer.
Collapse
Affiliation(s)
| | - Tsung-Ying Yang
- Department of Internal Medicine, Division of Chest Medicine, Taichung Veterans General Hospital, Taichung; Faculty of Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Ho Lin
- Department of Life Sciences; Program in Translational Medicine and Rong Hsing Research Center for Translational Medicine, National Chung Hsing University, Taichung, Taiwan
| | - Mei-Chih Chen
- Department of Nursing, Asia University; Translational Cell Therapy Center, Department of Medical Research, China Medical University Hospital, Taichung, Taiwan
| |
Collapse
|
25
|
Duan S, Li J, Tian J, Yin H, Zhai Q, Wu Y, Yao S, Zhang L. Crosstalk between let-7a-5p and BCL-xL in the Initiation of Toxic Autophagy in Lung Cancer. MOLECULAR THERAPY-ONCOLYTICS 2019; 15:69-78. [PMID: 31650027 PMCID: PMC6804504 DOI: 10.1016/j.omto.2019.08.010] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Accepted: 08/31/2019] [Indexed: 02/09/2023]
Abstract
Autophagy is essential for cellular metabolism and plays pivotal roles in carcinogenesis, while excessive autophagy induces toxicity and cell death. Our previous studies have suggested that let-7a-5p/BCL-xL might regulate autophagy in lung cancer, but the regulatory mechanism is unclear. The central goal of the study was to figure out the role of let-7a-5p/BCL-xL in the initiation of autophagy and its effect on the migration, invasion, and proliferation of A549 cells as well as its therapeutic potential in lung cancer. Based on the genome-wide expression profiles of lung cancer, BCL-xL and let-7a-5p were found to be dysregulated and negatively correlated in lung adenocarcinoma, which was associated with the survival of lung cancer. The crosstalk between BCL-xL and let-7a-5p was then investigated using dual-luciferase reporter assay, and it was found to suppress the migration and invasion of A549 cells. Further, we found that the crosstalk between BCL-xL and let-7a-5p could lead to toxic autophagy and cell death through activating the PI3K-signaling pathway, which was independent of apoptosis or pyroptosis. These findings indicate that let-7a-5p is a sensitive initiator for toxic autophagy in A549 lung cancer cells and is an appealing target for lung cancer therapy.
Collapse
Affiliation(s)
- Shuyin Duan
- School of Public Health, Zhengzhou University, Zhengzhou 450001, China
| | - Junxia Li
- School of Public Health and Management, Weifang Medical University, Weifang 261053, China
| | - Jiaqi Tian
- School of Public Health and Management, Weifang Medical University, Weifang 261053, China
| | - Haoyu Yin
- School of Public Health and Management, Weifang Medical University, Weifang 261053, China
| | - Qingfeng Zhai
- School of Public Health and Management, Weifang Medical University, Weifang 261053, China
| | - Yongjun Wu
- School of Public Health, Zhengzhou University, Zhengzhou 450001, China
| | - Sanqiao Yao
- School of Public Health, Xinxiang Medical University, Xinxiang 453000, China
| | - Lin Zhang
- Key Laboratory of Birth Regulation and Control Technology of National Health Commission of China, Shandong Maternal and Child Health Care Hospital, Jinan 250001, China
| |
Collapse
|