1
|
Yue Y, An G, Cao S, Li X, Du L, Xu D, Liu L. PLEKHA4 knockdown induces apoptosis in melanoma cells through the MAPK and β‑catenin signaling pathways. Mol Med Rep 2025; 31:99. [PMID: 39981886 DOI: 10.3892/mmr.2025.13464] [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: 09/28/2024] [Accepted: 01/15/2025] [Indexed: 02/22/2025] Open
Abstract
Malignant melanoma (MM) is a highly aggressive subtype of skin cancer characterized by a poor prognosis, particularly in the advanced stages. Despite advancements in targeted therapy and immunotherapy, the survival rates for MM remain low, underscoring the need for new therapeutic targets. Pleckstrin homology domain‑containing family A member 4 (PLEKHA4), which has regulatory functions in pivotal cellular processes, has emerged as a potential target in melanoma. The present study aimed to investigate the role of PLEKHA4 in melanoma progression, focusing on its influence on the MAPK and Wnt/β‑catenin signaling pathways. Bioinformatics analysis revealed that PLEKHA4 was upregulated in melanoma tissues, whereas PLEKHA4 knockdown in melanoma cell lines (A375 and A2058) significantly inhibited cell proliferation and migration, enhanced apoptosis and inhibited tumor growth in vivo. Mechanistic studies demonstrated that PLEKHA4 may exert its effects by modulating the MAPK signaling pathway through interactions with key proteins, including ERK, JNK and MEK. Additionally, PLEKHA4 was shown to impact apoptosis by regulating caspase‑3, COX2 and p65. Additionally, β‑catenin nuclear translocation was affected via the Wnt pathway. Moreover, PLEKHA4 knockdown reduced cMyc ubiquitination, consequently promoting its degradation. The present findings suggested that PLEKHA4 could promote melanoma cell proliferation by regulating both the MAPK and Wnt/β‑catenin pathways, thereby proposing PLEKHA4 as a promising therapeutic target for MM. Further studies are warranted to elucidate the mechanisms underlying PLEKHA4‑mediated modulation of cMyc ubiquitination.
Collapse
Affiliation(s)
- Yuyang Yue
- Department of Pathology, Yanbian University Hospital, Yanji, Jilin 133000, P.R. China
| | - Guangqi An
- Department of Ophthalmology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Shuxia Cao
- Key Laboratory of Cellular Function and Pharmacology of Jilin Province, Medical College of Yanbian University, Yanji, Jilin 133002, P.R. China
| | - Xiangdan Li
- Center of Morphological Experiment, Medical College of Yanbian University, Yanji, Jilin 133002, P.R. China
| | - Liping Du
- Department of Ophthalmology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Dongyuan Xu
- Key Laboratory of Cellular Function and Pharmacology of Jilin Province, Medical College of Yanbian University, Yanji, Jilin 133002, P.R. China
| | - Lan Liu
- Department of Pathology, Yanbian University Hospital, Yanji, Jilin 133000, P.R. China
| |
Collapse
|
2
|
Guo C, Kumar A, Liao C. Pterostilbene Exhibited the Anticancer Effect Against 1, 2-Dimethylhydrazine (DMH)-Induced Colorectal Cancer via Alteration of Oxidative Stress, Inflammation and Gut Microbiota. J Biochem Mol Toxicol 2025; 39:e70123. [PMID: 40084940 DOI: 10.1002/jbt.70123] [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: 10/16/2024] [Revised: 12/06/2024] [Accepted: 12/21/2024] [Indexed: 03/16/2025]
Abstract
Colorectal cancer (CRC) is the third leading cause of cancer-related deaths worldwide. The current investigation aimed to assess the chemoprotective effects of pterostilbene against 1,2-dimethylhydrazine (DMH)-induced colorectal cancer in mice. An in-silico study was conducted to perform docking studies against nuclear factor kappa factor (NF-κB). CRC was induced in mice by administering DMH (20 mg/kg) subcutaneously, and the mice were subsequently administered various dosages of pterostilbene (5, 10, and 15 mg/kg). At the end of the study, various biochemical parameters, including inflammatory cytokines, inflammatory markers, and antioxidant enzymes, were examined. Additionally, the mice's stools were collected for the analysis of intestinal microbiota. A total of 5 hydrogen bonds were identified between NF-κB and pterostilbene using LigPlot+. Pterostilbene significantly (p < 0.001) reduced tumor incidence, tumor weight, and increased body weight. Pterostilbene significantly (p < 0.001) altered the levels of lipid peroxidation, reduced glutathione, superoxide dismutase, glutathione peroxidase, and catalase as well as the activity of both phase I and phase II enzymes. Furthermore, pterostilbene significantly (p < 0.001) decreased the levels of proinflammatory cytokines such as tumor necrosis factor-α, interleukin-6, interferon-γ, and interleukin-1β, while increasing the levels of anti-inflammatory cytokines like interleukin-4 and interleukin-10. Pterostilbene considerably suppressed the levels of cyclooxygenase-2 and prostaglandin E2, as well as inducible nitric oxide synthase and simultaneously elevated the levels of apoptosis-related parameters, including caspase-3, caspase-8, and caspase-9. Moreover, pterostilbene significantly reduced the abundance of Staphylococcus in the intestinal microbiota and enhanced the levels of beneficial bacteria, such as Bifidobacterium, Akkermansia, and Lactobacillus. Pterostilbene demonstrated a chemoprotective effect against CRC by effectively reducing oxidative stress, mitigating inflammatory responses, and inducing alterations in gut microbiota levels.
Collapse
Affiliation(s)
- Chengyun Guo
- Department of Anorectal, Sanmenxia Central Hospital, Henan, China
| | - Ankit Kumar
- Department of Pharmacology, Venkateshwara College of Pharmacy, Meerut, India
| | - Chao Liao
- Department of Anorectal, Ankang Hospital of Traditional Chinese Medicine, Ankang, China
| |
Collapse
|
3
|
Shoari A, Ashja Ardalan A, Dimesa AM, Coban MA. Targeting Invasion: The Role of MMP-2 and MMP-9 Inhibition in Colorectal Cancer Therapy. Biomolecules 2024; 15:35. [PMID: 39858430 PMCID: PMC11762759 DOI: 10.3390/biom15010035] [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: 12/09/2024] [Revised: 12/27/2024] [Accepted: 12/29/2024] [Indexed: 01/27/2025] Open
Abstract
Colorectal cancer (CRC) remains one of the most prevalent and lethal cancers worldwide, prompting ongoing research into innovative therapeutic strategies. This review aims to systematically evaluate the role of gelatinases, specifically MMP-2 and MMP-9, as therapeutic targets in CRC, providing a critical analysis of their potential to improve patient outcomes. Gelatinases, specifically MMP-2 and MMP-9, play critical roles in the processes of tumor growth, invasion, and metastasis. Their expression and activity are significantly elevated in CRC, correlating with poor prognosis and lower survival rates. This review provides a comprehensive overview of the pathophysiological roles of gelatinases in CRC, highlighting their contribution to tumor microenvironment modulation, angiogenesis, and the metastatic cascade. We also critically evaluate recent advancements in the development of gelatinase inhibitors, including small molecule inhibitors, natural compounds, and novel therapeutic approaches like gene silencing techniques. Challenges such as nonspecificity, adverse side effects, and resistance mechanisms are discussed. We explore the potential of gelatinase inhibition in combination therapies, particularly with conventional chemotherapy and emerging targeted treatments, to enhance therapeutic efficacy and overcome resistance. The novelty of this review lies in its integration of recent findings on diverse inhibition strategies with insights into their clinical relevance, offering a roadmap for future research. By addressing the limitations of current approaches and proposing novel strategies, this review underscores the potential of gelatinase inhibitors in CRC prevention and therapy, inspiring further exploration in this promising area of oncological treatment.
Collapse
Affiliation(s)
- Alireza Shoari
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL 32224, USA;
| | - Arghavan Ashja Ardalan
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum, University of Bologna, 40126 Bologna, Italy;
| | | | - Mathew A. Coban
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL 32224, USA;
| |
Collapse
|
4
|
Yang B, Zheng G, Lu F. PDCL3 as a prognostic factor and associated with the VEGF signaling pathway in glioma. J Gene Med 2024; 26:e3724. [PMID: 39107869 DOI: 10.1002/jgm.3724] [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: 05/20/2024] [Revised: 06/29/2024] [Accepted: 07/16/2024] [Indexed: 09/14/2024] Open
Abstract
BACKGROUND New targeted drugs about angiogenesis could develop the treatment of glioma. We aimed to explore the role of phosducin like 3 (PDCL3) in angiogenesis of glioma. MATERIALS AND METHODS RNA sequencing data and matched clinical data were downloaded from The Cancer Genome Atlas (TCGA) and Chinese Glioma Genome Atlas (CGGA) databases. To screen for the reliable genes with the filtering analyses, survival, multivariate Cox, receiver operating characteristic (ROC) curve filtration, and clinical correlation analyses were performed. The PDCL3 gene was validated by immunohistochemistry as a reliable gene for further analysis. Then we used the combined data of TCGA and Genotype-Tissue Expression from UCSC to detect the differential gene expression of PDCL3. Related signal pathways in glioma were explored by the gene set enrichment analysis and co-expression analysis. Lastly, we performed in vitro experiments to verify the gene functions and related mechanisms. RESULTS The three filtering analyses and immunostaining indicated that the expression of PDCL3 in glioma tissues was higher than the normal tissues. Gene function analysis showed that PDCL3 activated the vascular endothelial growth factor (VEGF) signal pathway, and its mechanism was related to pathways in cancer, like NOD like receptor signaling pathway, the RIG-I like receptor signaling pathway and the P53 signaling pathway by MAPK/AKT in gliomas. This suggested that the proliferation, migration and invasion of glioma cells might be inhibited by the downregulation of PDCL3 in vitro, which may be related to the activation of VEGF signaling pathway. CONCLUSION We demonstrated that PDCL3 could function as an independent adverse prognostic marker in glioma. Its pro-oncogenic mechanism may be related to the VEGF signaling pathway.
Collapse
Affiliation(s)
- Bo Yang
- Department of Neurosurgery, Shengli Clinical Medical College of Fujian Medical University, Fuzhou University Affiliated Provincial Hospital, Fujian Provincial Hospital, Fuzhou, China
| | - Guangwei Zheng
- Department of Neurosurgery, Shengli Clinical Medical College of Fujian Medical University, Fuzhou University Affiliated Provincial Hospital, Fujian Provincial Hospital, Fuzhou, China
| | - Feng Lu
- Department of Neurosurgery, Shengli Clinical Medical College of Fujian Medical University, Fuzhou University Affiliated Provincial Hospital, Fujian Provincial Hospital, Fuzhou, China
| |
Collapse
|
5
|
Yu Z, Peng Y, Gao J, Zhou M, Shi L, Zhao F, Wang C, Tian X, Feng L, Huo X, Zhang B, Liu M, Fang D, Ma X. The p23 co-chaperone is a succinate-activated COX-2 transcription factor in lung adenocarcinoma tumorigenesis. SCIENCE ADVANCES 2023; 9:eade0387. [PMID: 37390202 PMCID: PMC10313168 DOI: 10.1126/sciadv.ade0387] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Accepted: 05/30/2023] [Indexed: 07/02/2023]
Abstract
P23, historically known as a heat shock protein 90 (HSP90) co-chaperone, exerts some of its critical functions in an HSP90-independent manner, particularly when it translocates into the nucleus. The molecular nature underlying how this HSP90-independent p23 function is achieved remains as a biological mystery. Here, we found that p23 is a previously unidentified transcription factor of COX-2, and its nuclear localization predicts the poor clinical outcomes. Intratumor succinate promotes p23 succinylation at K7, K33, and K79, which drives its nuclear translocation for COX-2 transcription and consequently fascinates tumor growth. We then identified M16 as a potent p23 succinylation inhibitor from 1.6 million compounds through a combined virtual and biological screening. M16 inhibited p23 succinylation and nuclear translocation, attenuated COX-2 transcription in a p23-dependent manner, and markedly suppressed tumor growth. Therefore, our study defines p23 as a succinate-activated transcription factor in tumor progression and provides a rationale for inhibiting p23 succinylation as an anticancer chemotherapy.
Collapse
Affiliation(s)
- Zhenlong Yu
- College of Pharmacy, the Second Affiliated Hospital, Dalian Medical University, Dalian 116000, China
| | - Yulin Peng
- College of Pharmacy, the Second Affiliated Hospital, Dalian Medical University, Dalian 116000, China
| | - Jian Gao
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou 221004, China
- School of Medicine, Anhui University of Science and Technology, Huainan 232001, China
| | - Meirong Zhou
- College of Pharmacy, the Second Affiliated Hospital, Dalian Medical University, Dalian 116000, China
| | - Lei Shi
- College of Pharmacy, the Second Affiliated Hospital, Dalian Medical University, Dalian 116000, China
| | - Feng Zhao
- College of Pharmacy, the Second Affiliated Hospital, Dalian Medical University, Dalian 116000, China
| | - Chao Wang
- College of Pharmacy, the Second Affiliated Hospital, Dalian Medical University, Dalian 116000, China
| | - Xiangge Tian
- College of Pharmacy, the Second Affiliated Hospital, Dalian Medical University, Dalian 116000, China
| | - Lei Feng
- College of Pharmacy, the Second Affiliated Hospital, Dalian Medical University, Dalian 116000, China
| | - Xiaokui Huo
- College of Pharmacy, the Second Affiliated Hospital, Dalian Medical University, Dalian 116000, China
| | - Baojing Zhang
- College of Pharmacy, the Second Affiliated Hospital, Dalian Medical University, Dalian 116000, China
| | - Min Liu
- Neurology Department, Dalian University Affiliated Xinhua Hospital, Dalian 116021, China
| | - Deyu Fang
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Xiaochi Ma
- College of Pharmacy, the Second Affiliated Hospital, Dalian Medical University, Dalian 116000, China
| |
Collapse
|
6
|
Ononitol Monohydrate-A Glycoside Potentially Inhibit HT-115 Human Colorectal Cancer Cell Proliferation through COX-2/PGE-2 Inflammatory Axis Regulations. Int J Mol Sci 2022; 23:ijms232214440. [PMID: 36430918 PMCID: PMC9696259 DOI: 10.3390/ijms232214440] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 11/08/2022] [Accepted: 11/10/2022] [Indexed: 11/22/2022] Open
Abstract
We aimed to inhibit HT-115 human colorectal cancer cell proliferation using ononitol monohydrate (OMH), a bioactive principle isolated from Cassia tora (L.). The cytotoxicity of OMH has been assayed using MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide), cell and nuclear morphology, and apoptosis mechanisms have been analyzed using real-time PCR. Higher doses of OMH potentially inhibit 84% of HT-115 cell viability; we observed that the IC50 level was 3.2 µM in 24 h and 1.5 µM in 48 h. The treatment with 3.2 µM of OMH for 48 h characteristically showed 64% apoptotic cells and 3% necrotic cells, confirmed by propidium iodide and acridine orange/ethidium bromide (AO/ErBr) staining. We found the overexpression of cyclooxygenase-2 (COX-2) and prostaglandin E2 (PGE-2) in the control HT-115 cells, which was directly associated with colorectal tumorigenesis. However, 3.2 µM of OMH treatment to HT-115 cells for 48 h significantly reduced inflammatory genes, such as TNF-α/IL-1β and COX-2/PGE-2. The downregulation of COX-2 and PGE-2 was more significant with the 3.2 µM dose when compared to the 1.5 µM dose of OMH. Additionally, the protein levels of COX-2 and PGE-2 were decreased in the 3.2 µM OMH-treated cells compared to the control. We found significantly (p ≤ 0.01) increased mRNA expression levels of tumor-suppressor genes, such as pRb2, Cdkn1a, p53, and caspase-3, and decreased Bcl-2, mdm2, and PCNA after 48 h was confirmed with apoptotic stimulation. In conclusion, the antiproliferative effect of OMH via the early suppression of protumorigenic inflammatory agents TNF-α/IL-1β, COX-2/PGE-2 expression, and the increased expression levels of tumor-suppressor genes Cdkn1a and pRb2, which enhanced the activation of Bax and p53.
Collapse
|
7
|
Chantawong P, Mamom T, Tangtrongsup S, Chitsanoor S, Boonsriroj H. First study on the immunohistochemical expression of cyclooxygenase-2 and clinicopathological association in canine hepatoid gland neoplasms. Vet World 2022; 15:2432-2441. [DOI: 10.14202/vetworld.2022.2432-2441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Accepted: 09/01/2022] [Indexed: 11/16/2022] Open
Abstract
Background and Aim: Hepatoid gland neoplasms (HGNs) constitute one of the most common cutaneous tumors that arise from perianal glands in dogs and are clinically characterized by rapid growth. Cyclooxygenase-2 (COX-2), the inducible form of the enzyme, is associated with several hallmarks of tumorigenesis. Its expression has been confirmed in several human and animal neoplastic tissues, but there are no reports in hepatoid gland tissues. Therefore, this study aimed to investigate COX-2 immunoexpression in canine HGNs, compare the expression among groups of normal hepatoid glands, hepatoid gland adenomas (HGAs), hepatoid gland epitheliomas (HGEs), and hepatoid gland carcinomas (HGCs), and assess the association of the COX-2 expression with clinicopathological features.
Materials and Methods: Sixty-one formalin-fixed paraffin-embedded canine hepatoid gland tissues (20 samples of HGAs, 16 of HGEs, 15 of HGCs, and 10 of normal hepatoid glands) were analyzed for COX-2 expression using immunohistochemistry with scoring for percentage positivity and intensity. Multiple comparisons of COX-2 expression among normal and neoplastic hepatoid glands and the associations between COX-2 expression and clinicopathological features were analyzed.
Results: Cyclooxygenase-2 expression was not detected in 60% of normal hepatoid glands and 25% of HGAs. Seventy-five percent of HGAs had a weak expression, while 43.7% and 56.3% of HGEs showed weak and moderate expression, respectively. The expression of HGCs ranged from weak (13.3%) to moderate (33.3%) and strong (53.3%). The immunoreactivity score of COX-2 labeling was significantly different among the normal and neoplastic hepatoid glands (p < 0.0001). The highest score was observed in the HGCs. Only in HGCs, the strong COX-2 expression was significantly associated with some clinicopathological features, including tissue invasion (p = 0.007) and necrosis (p = 0.029).
Conclusion: These results suggest that COX-2 may play a role in the modulation of neoplastic cell growth. These preliminary data lead to further investigation on the potential of COX-2 expression as a prognostic indicator and COX-2 inhibitors for canine HGCs treatment.
Collapse
Affiliation(s)
- Pinkarn Chantawong
- Department of Companion Animal and Wildlife Clinic, Faculty of Veterinary Medicine, Chiang Mai University, Chiang Mai 50100, Thailand; Integrative Research Center for Veterinary Preventive Medicine, Faculty of Veterinary Medicine, Chiang Mai University, Chiang Mai 50100, Thailand
| | - Thanongsak Mamom
- Department of Veterinary Pathology, Faculty of Veterinary Medicine, Mahanakorn University of Technology, Bangkok 10530, Thailand; Mahanakorn Veterinary Diagnostic Center, Faculty of Veterinary Medicine, Mahanakorn University of Technology, Bangkok 10530, Thailand
| | - Sahatchai Tangtrongsup
- Department of Companion Animal and Wildlife Clinic, Faculty of Veterinary Medicine, Chiang Mai University, Chiang Mai 50100, Thailand; Research Center of Producing and Development of Products and Innovations for Animal Health and Production, Chiang Mai University, Chiang Mai 50100, Thailand
| | - Setthakit Chitsanoor
- Department of Veterinary Pathology, Faculty of Veterinary Medicine, Mahanakorn University of Technology, Bangkok 10530, Thailand; Mahanakorn Veterinary Diagnostic Center, Faculty of Veterinary Medicine, Mahanakorn University of Technology, Bangkok 10530, Thailand
| | - Hassadin Boonsriroj
- Department of Veterinary Pathology, Faculty of Veterinary Medicine, Mahanakorn University of Technology, Bangkok 10530, Thailand; Mahanakorn Veterinary Diagnostic Center, Faculty of Veterinary Medicine, Mahanakorn University of Technology, Bangkok 10530, Thailand
| |
Collapse
|
8
|
Zafari N, Khosravi F, Rezaee Z, Esfandyari S, Bahiraei M, Bahramy A, Ferns GA, Avan A. The role of the tumor microenvironment in colorectal cancer and the potential therapeutic approaches. J Clin Lab Anal 2022; 36:e24585. [PMID: 35808903 PMCID: PMC9396196 DOI: 10.1002/jcla.24585] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 06/07/2022] [Accepted: 06/23/2022] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND Colorectal cancer (CRC) with a high prevalence is recognized as the fourth most common cause of cancer-related death globally. Over the past decade, there has been growing interest in the network of tumor cells, stromal cells, immune cells, blood vessel cells, and fibroblasts that comprise the tumor microenvironment (TME) to identify new therapeutic interventions. METHODS Databases, such as Google Scholar, PubMed, and Scopus, were searched to provide an overview of the recent research progress related to targeting the TME as a novel therapeutic approach. RESULTS Tumor microenvironment as a result of the cross talk between these cells may result in either advantages or disadvantages in tumor development and metastasis, affecting the signals and responses from the surrounding cells. Whilst chemotherapy has led to an improvement in CRC patients' survival, the metastatic aspect of the disease remains difficult to avoid. CONCLUSIONS The present review emphasizes the structure and function of the TME, alterations in the TME, its role in the incidence and progression of CRC, the effects on tumor development and metastasis, and also the potential of its alterations as therapeutic targets. It should be noted that providing novel studies in this field of research might help us to achieve practical therapeutic strategies based on their interaction.
Collapse
Affiliation(s)
- Narges Zafari
- Department of Medical Genetics, School of MedicineTehran University of Medical SciencesTehranIran
| | - Fatemeh Khosravi
- Molecular Medicine Research Center, Hormozgan Health InstituteHormozgan University of Medical SciencesBandar AbbasIran
| | - Zahra Rezaee
- Department of Medical Genetics, Faculty of Medical SciencesTarbiat Modares UniversityTehranIran
| | - Sahar Esfandyari
- Department of Anatomy, School of MedicineTehran University of Medical SciencesTehranIran
| | - Mohamad Bahiraei
- Department of Radiology, Besat HospitalHamedan University of Medical SciencesHamedanIran
| | - Afshin Bahramy
- Department of Medical Genetics, Faculty of Medical SciencesTarbiat Modares UniversityTehranIran
| | - Gordon A. Ferns
- Brighton & Sussex Medical SchoolDivision of Medical EducationSussexUK
| | - Amir Avan
- Metabolic Syndrome Research CenterMashhad University of Medical SciencesMashhadIran
- Basic Medical Sciences InstituteMashhad University of Medical SciencesMashhadIran
- Medical Genetics Research CenterMashhad University of Medical SciencesMashhadIran
| |
Collapse
|
9
|
Meloxicam Inhibits Hepatocellular Carcinoma Progression and Enhances the Sensitivity of Immunotherapy via the MicroRNA-200/PD-L1 Pathway. JOURNAL OF ONCOLOGY 2022; 2022:4598573. [PMID: 35237322 PMCID: PMC8885196 DOI: 10.1155/2022/4598573] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Revised: 01/12/2022] [Accepted: 01/28/2022] [Indexed: 12/14/2022]
Abstract
Background. Hepatocellular carcinoma (HCC) has become the sixth most common cancer and the third leading cause of cancer death in the world. Although the research achievements of tumor immunotherapy have made great progress, especially the combination of immune targeted therapy has achieved good curative effect in HCC, but only a few patients are suitable for it and benefit from it. Therefore, there is an urgent need to find new effective drugs to treat HCC or to enhance the sensitivity of immunotherapy. Methods. Meloxicam, a COX2 inhibitor with strong anti-HCC potential, was screened from 800 small molecules approved by FDA. The effect of meloxicam on the proliferation, invasion, and migration of HCC cell lines was evaluated by cell phenotype analysis. The Human Protein Atlas database and the TISCH database were used to analyze COX2 data in single cells, and the TISIDB database was used to analyze the correlation of COX2 with immune function. The real-time quantitative polymerase chain reaction (qRT-PCR) and western blot were used to evaluate the level of PD-L1 and CD155 in HCC cell lines treated with meloxicam and further explore its possible mechanism. In vivo experiments were applied to verify the effect of meloxicam combined with anti-PD1 therapy on HCC tumor growth in mice. Results. Meloxicam can significantly inhibit the proliferation, invasion, and migration of HCC cells. The TISIDB database indicated that the COX2 was strongly associated with immunoinhibitors and immunostimulators. Meloxicam upregulated the level of PD-L1 in HCC cell lines and animal models. In terms of mechanism, meloxicam inhibited microRNA-200, thereby upregulating PD-L1. In vitro experiments showed that both meloxicam and anti-PD1 had inhibitory effects on the growth of HCC tumors. Compared with meloxicam and anti-PD1 alone, the combination therapy showed stronger antitumor properties. Immunohistochemical analysis confirmed that meloxicam enhanced the antitumor immune activity in the tumor microenvironment. Conclusion. Our study showed meloxicam inhibited HCC progression and enhanced the sensitivity of immunotherapy via the microRNA-200/PD-L1 pathway.
Collapse
|
10
|
Cárdenas DM, Gómez Rave LJ, Soto JA. Biological Activity of Sacha Inchi ( Plukenetia volubilis Linneo) and Potential Uses in Human Health: A Review. Food Technol Biotechnol 2021; 59:253-266. [PMID: 34759758 PMCID: PMC8542186 DOI: 10.17113/ftb.59.03.21.6683] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 07/13/2021] [Indexed: 11/20/2022] Open
Abstract
Sacha inchi (Plukenetia volubilis Linneo) is an ancestral plant originating in the Amazon jungle that has been adopted as a food source due to its high nutritional value, which has gradually been recognized to have potential benefits for human health. Diverse prospective studies have evaluated the effect of consuming components from the plant, derivatives from its seeds, leaves and shell on preventing the risk of cardiovascular disease, chronic inflammatory disease, dermatitis and controlling tumor proliferation, especially given its recognized high content of essential fatty acids, phenolic compounds and vitamin E, showing antioxidant, hypolipidemic, immunomodulation and emollient activity, as well as the capacity to remove heavy metals from aqueous solutions. This review offers a complete description of the existing information on the use and biological activity of P. volubilis L., based on its essential lipid components and evidenced on its use in the field of human health, in prevention, therapeutic and nutritional contexts, along with industrial uses, making it a promising bioresource.
Collapse
Affiliation(s)
- Denny M Cárdenas
- Universidad de Santander, Facultad de Ciencias Médicas y de la Salud, Grupo de Investigación BIOGEN, Avenida 4 calle 10N-61, 540001 Cúcuta, Colombia
| | - Lyz Jenny Gómez Rave
- Institución Universitaria Colegio Mayor de Antioquia, Facultad de Ciencias de la Salud, Masira Research Institute, Calle 70 No. 55-210, Bucaramanga, Colombia
| | - Javier Andrés Soto
- Universidad de Santander, Facultad de Ciencias Médicas y de la Salud, Grupo de Investigación BIOGEN, Avenida 4 calle 10N-61, 540001 Cúcuta, Colombia
| |
Collapse
|
11
|
Mani S, Swargiary G, Ralph SJ. Targeting the redox imbalance in mitochondria: A novel mode for cancer therapy. Mitochondrion 2021; 62:50-73. [PMID: 34758363 DOI: 10.1016/j.mito.2021.11.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 10/14/2021] [Accepted: 11/01/2021] [Indexed: 12/19/2022]
Abstract
Changes in reactive oxygen species (ROS) levels affect many aspects of cell behavior. During carcinogenesis, moderate ROS production modifies gene expression to alter cell function, elevating metabolic activity and ROS. To avoid extreme ROS-activated death, cancer cells increase antioxidative capacity, regulating sustained ROS levels that promote growth. Anticancer therapies are exploring inducing supranormal, cytotoxic oxidative stress levels either inhibiting antioxidative capacity or promoting excess ROS to selectively destroy cancer cells, triggering mechanisms such as apoptosis, autophagy, necrosis, or ferroptosis. This review exemplifies pro-oxidants (natural/synthetic/repurposed drugs) and their clinical significance as cancer therapies providing revolutionary approaches.
Collapse
Affiliation(s)
- Shalini Mani
- Centre for Emerging Diseases, Department of Biotechnology, Jaypee Institute of Information Technology, Noida, India.
| | - Geeta Swargiary
- Centre for Emerging Diseases, Department of Biotechnology, Jaypee Institute of Information Technology, Noida, India
| | - Stephen J Ralph
- School of Medical Science, Griffith University, Southport, Australia.
| |
Collapse
|
12
|
Qin J, Li H, Yu W, Wei L, Wen B. Effect of cold exposure and capsaicin on the expression of histone acetylation and Toll-like receptors in 1,2-dimethylhydrazine-induced colon carcinogenesis. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2021; 28:60981-60992. [PMID: 34165751 DOI: 10.1007/s11356-021-14849-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 06/08/2021] [Indexed: 06/13/2023]
Abstract
Previous studies have indicated that capsaicin-rich diet and cold weather have shown strong association with tumor incidence. Thus, we investigated the effects of capsaicin and cold exposure in 1,2-dimethylhydrazine (DMH)-induced colorectal cancer as well as the mechanisms underlying capsaicin and cold-induced CRC. Rats were randomly divided into four groups and received cold still water and capsaicin via intragastric gavage until the end of the experiment. The rat's body weight, thymus weight, and food intakes were assessed. Global levels of histone H3K9, H3K18, H3K27, and H4K16 acetylation and histone deacetylase (HDACs) in colon mucosa were assessed by western blot. Expression levels of Toll-like receptors 2 (TLR2) and Toll-like receptors 4 (TLR4) were measured by western blot and reverse-transcriptase quantitative polymerase chain reaction (qPCR). We found that cold and low-dose capsaicin increased tumor numbers and multiplicity, although there were no differences in tumor incidence. Additionally, rat exposure to cold water and capsaicin display further higher levels of histone H3 lysine 9 (H3K9AC), histone H3 lysine 18 (H3K18AC), histone H3 lysine 27 (H3K27AC), and HDACs compared with the DMH and normal rats. In contrast, a considerable decrease of histone H4 lysine 16 (H4K16AC) was detected in the colon mucosa. Cold and low-dose capsaicin exposure groups were also increased TLR2 and TLR4 protein levels and mRNA levels. These results suggest that chronic cold exposure and capsaicin at a low-dose intervention exacerbate ectopic expression of global histone acetylation and TLR level, which are crucial mechanisms responsible for the progression of colorectal cancer in rats.
Collapse
Affiliation(s)
- Jingchun Qin
- Institute of Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510000, China
| | - Huixuan Li
- National Center for Respiratory Medicine, State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Weitao Yu
- The Second People's Hospital Lianyungang, Lianyungang, China
| | - Li Wei
- Liuzhou Municipal Liutie Central Hospital, Liuzhou, China
| | - Bin Wen
- Institute of Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510000, China.
| |
Collapse
|
13
|
Qin JC, Yu WT, Li HX, Liang YQ, Nong FF, Wen B. Cold exposure and capsaicin promote 1,2-dimethylhyrazine-induced colon carcinogenesis in rats correlates with extracellular matrix remodeling. World J Gastroenterol 2021; 27:6615-6630. [PMID: 34754156 PMCID: PMC8554402 DOI: 10.3748/wjg.v27.i39.6615] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 07/02/2021] [Accepted: 07/30/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Extracellular matrix (ECM) remodeling and stiffening, which are correlated with tumor malignancy, drives tumor development. However, the relationship between ECM remodeling and rat experimental model of 1,2-dimethylhyrazine (DMH)-induced colorectal cancer (CRC) imposed by cold and capsaicin exposure remains unclear.
AIM To explore the effects of cold exposure and capsaicin on ECM remodeling and ECM enzymes in DMH-induced CRC.
METHODS For histopathological analysis, the sections of colon tissues were stained with hematoxylin and eosin, Masson’s trichrome, Picrosirius red, and Weigert’s Resorcin-Fuchsin to observe the remodeling of collagen and elastin. Additionally, the protein expression level of type I collagen (COL I), type 3 collagen (COL III0, elastin, matrix metalloproteinase (MMP) 1, MMP2, MMP9, and tissue-specific matrix metalloproteinase 1 (TIMP1) was assessed by immunohistochemistry. The messenger RNA (mRNA) levels of COL I, COL III, elastin, and lysyl oxidase-like-2 (LOXL2) in the colon tissues of rats was measured by reverse-transcriptase quantitative polymerase chain reaction.
RESULTS Although no differences were observed in the proportion of adenomas, a trend towards the increase of invasive tumors was observed in the cold and capsaicin group. The cold exposure group had a metastasis rate compared with the other groups. Additionally, abnormal accumulation of both collagen and elastin was observed in the cold exposure and capsaicin group. Specifically, collagen quantitative analysis showed increased length, width, angle, and straightness compared with the DMH group. Collagen deposition and straightness were significantly increased in the cold exposure group compared with the capsaicin group. Cold exposure and capsaicin significantly increased the protein levels of COL I, elastin, and LOXL2 along with increases in their mRNA levels in the colon tissues compared with the DMH group, while COL III did not show a significant difference. Furthermore, in immunohistochemical evaluations, MMP1, MMP2, MMP9, and TIMP1 staining increased in the cold exposure and capsaicin group compared with the DMH group.
CONCLUSION These results suggest that chronic cold and capsaicin exposure further increased the deposition of collagen and elastin in the colonic tissue. Increased COL I and elastin mRNA and protein levels expression may account for the enhanced ECM remodel and stiffness variations of colon tissue. The upregulated expression of the LOXL2 and physiological imbalance between MMP/TIMP activation and deactivation could contribute to the progression of the CRC resulting from cold and capsaicin exposure.
Collapse
Affiliation(s)
- Jing-Chun Qin
- Institute of Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangdong 530001, Guangdong Province, China
- Liuzhou People’s Hospital, Guangxi, 545006, Guangxi Province China
| | - Wei-Tao Yu
- Traditional Chinese Medicine Department, The Second People’s Hospital of Lianyungang, Lianyungang 222000, Jiangsu Province, China
| | - Hui-Xuan Li
- National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, State Key Laboratory of Respiratory Disease and National Clinical Research Center for Respiratory Disease, Guangdong 510000, Guangdong Province, China
| | - Yu-Qi Liang
- Institute of Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangdong 530001, Guangdong Province, China
| | - Fei-Fei Nong
- Institute of Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangdong 530001, Guangdong Province, China
| | - Bin Wen
- Institute of Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangdong 530001, Guangdong Province, China
| |
Collapse
|
14
|
Tomita K, Nagasawa T, Kuwahara Y, Torii S, Igarashi K, Roudkenar MH, Roushandeh AM, Kurimasa A, Sato T. MiR-7-5p Is Involved in Ferroptosis Signaling and Radioresistance Thru the Generation of ROS in Radioresistant HeLa and SAS Cell Lines. Int J Mol Sci 2021; 22:8300. [PMID: 34361070 PMCID: PMC8348045 DOI: 10.3390/ijms22158300] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 07/29/2021] [Accepted: 07/30/2021] [Indexed: 12/11/2022] Open
Abstract
In cancer therapy, radioresistance or chemoresistance cells are major problems. We established clinically relevant radioresistant (CRR) cells that can survive over 30 days after 2 Gy/day X-ray exposures. These cells also show resistance to anticancer agents and hydrogen peroxide (H2O2). We have previously demonstrated that all the CRR cells examined had up-regulated miR-7-5p and after miR-7-5p knockdown, they lost radioresistance. However, the mechanism of losing radioresistance remains to be elucidated. Therefore, we investigated the role of miR-7-5p in radioresistance by knockdown of miR-7-5p using CRR cells. As a result, knockdown of miR-7-5p increased reactive oxygen species (ROS), mitochondrial membrane potential, and intracellular Fe2+ amount. Furthermore, miR-7-5p knockdown results in the down-regulation of the iron storage gene expression such as ferritin, up-regulation of the ferroptosis marker ALOX12 gene expression, and increases of Liperfluo amount. H2O2 treatment after ALOX12 overexpression led to the enhancement of intracellular H2O2 amount and lipid peroxidation. By contrast, miR-7-5p knockdown seemed not to be involved in COX-2 and glycolysis signaling but affected the morphology of CRR cells. These results indicate that miR-7-5p control radioresistance via ROS generation that leads to ferroptosis.
Collapse
Affiliation(s)
- Kazuo Tomita
- Department of Applied Pharmacology, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima-City 890-8544, Kagoshima, Japan; (T.N.); (Y.K.); (K.I.); (M.H.R.); (A.M.R.); (T.S.)
| | - Taisuke Nagasawa
- Department of Applied Pharmacology, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima-City 890-8544, Kagoshima, Japan; (T.N.); (Y.K.); (K.I.); (M.H.R.); (A.M.R.); (T.S.)
| | - Yoshikazu Kuwahara
- Department of Applied Pharmacology, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima-City 890-8544, Kagoshima, Japan; (T.N.); (Y.K.); (K.I.); (M.H.R.); (A.M.R.); (T.S.)
- Division of Radiation Biology and Medicine, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, Sendai-City 983-8536, Miyagi, Japan;
| | - Seiji Torii
- Center for Food Science and Wellness, Gunma University, Maebashi-City 371-8510, Gunma, Japan;
| | - Kento Igarashi
- Department of Applied Pharmacology, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima-City 890-8544, Kagoshima, Japan; (T.N.); (Y.K.); (K.I.); (M.H.R.); (A.M.R.); (T.S.)
| | - Mehryar Habibi Roudkenar
- Department of Applied Pharmacology, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima-City 890-8544, Kagoshima, Japan; (T.N.); (Y.K.); (K.I.); (M.H.R.); (A.M.R.); (T.S.)
- Burn and Regenerative Medicine Research Center, Velayat Hospital, School of Medicine, Guilan University of Medical Sciences, Rasht 41937-13194, Iran
| | - Amaneh Mohammadi Roushandeh
- Department of Applied Pharmacology, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima-City 890-8544, Kagoshima, Japan; (T.N.); (Y.K.); (K.I.); (M.H.R.); (A.M.R.); (T.S.)
- Burn and Regenerative Medicine Research Center, Velayat Hospital, School of Medicine, Guilan University of Medical Sciences, Rasht 41937-13194, Iran
| | - Akihiro Kurimasa
- Division of Radiation Biology and Medicine, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, Sendai-City 983-8536, Miyagi, Japan;
| | - Tomoaki Sato
- Department of Applied Pharmacology, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima-City 890-8544, Kagoshima, Japan; (T.N.); (Y.K.); (K.I.); (M.H.R.); (A.M.R.); (T.S.)
| |
Collapse
|
15
|
Khafaga AF, Shamma RN, Abdeen A, Barakat AM, Noreldin AE, Elzoghby AO, Sallam MA. Celecoxib repurposing in cancer therapy: molecular mechanisms and nanomedicine-based delivery technologies. Nanomedicine (Lond) 2021; 16:1691-1712. [PMID: 34264123 DOI: 10.2217/nnm-2021-0086] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
While cancer remains a significant global health problem, advances in cancer biology, deep understanding of its underlaying mechanism and identification of specific molecular targets allowed the development of new therapeutic options. Drug repurposing poses several advantages as reduced cost and better safety compared with new compounds development. COX-2 inhibitors are one of the most promising drug classes for repurposing in cancer therapy. In this review, we provide an overview of the detailed mechanism and rationale of COX-2 inhibitors as anticancer agents and we highlight the most promising research efforts on nanotechnological approaches to enhance COX-2 inhibitors delivery with special focus on celecoxib as the most widely studied agent for chemoprevention or combined with chemotherapeutic and herbal drugs for combating various cancers.
Collapse
Affiliation(s)
- Asmaa F Khafaga
- Department of Pathology, Faculty of Veterinary Medicine, Alexandria University, Edfina, 22758, Egypt
| | - Rehab N Shamma
- Department of Pharmaceutics & Industrial Pharmacy, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt
| | - Ahmed Abdeen
- Department of Forensic Medicine & Toxicology, Faculty of Veterinary Medicine, Benha University, Toukh, 13736, Egypt
| | | | - Ahmed E Noreldin
- Department of Histology & Cytology, Faculty of Veterinary Medicine, Damanhour University, Damanhour, 22516, Egypt
| | - Ahmed O Elzoghby
- Cancer Nanotechnology Research Laboratory (CNRL), Department of Industrial Pharmacy, Faculty of Pharmacy, Alexandria University, Alexandria, 21521, Egypt.,Department of Industrial Pharmacy, Faculty of Pharmacy, Alexandria University, Alexandria, 21521, Egypt
| | - Marwa A Sallam
- Department of Industrial Pharmacy, Faculty of Pharmacy, Alexandria University, Alexandria, 21521, Egypt
| |
Collapse
|
16
|
Wang X, Sheng W, Xu T, Xu J, Gao R, Zhang Z. CircRNA hsa_circ_0110102 inhibited macrophage activation and hepatocellular carcinoma progression via miR-580-5p/PPARα/CCL2 pathway. Aging (Albany NY) 2021; 13:11969-11987. [PMID: 33891564 PMCID: PMC8109088 DOI: 10.18632/aging.202900] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 03/02/2021] [Indexed: 04/26/2023]
Abstract
Circular RNAs (circRNAs) have critical regulatory roles in tumor biology. However, their contributions in hepatocellular carcinoma (HCC) still remain enigmatic. The present study aimed to investigate the molecular mechanisms underlying the involvement of hsa_circ_0110102 in the occurrence and development of HCC. The expression level of hsa_circ_0110102 was significantly downregulated in HCC cell lines and tissues, which was associated with poor prognosis. Knockdown hsa_circ_0110102 significantly promoted cell proliferation, migration, and invasion. Moreover, the interaction between hsa_circ_0110102 and miR-580-5p was predicted and verified by luciferase assay and RNA pull-down. The findings indicated that hsa_circ_0110102 functioned as a sponge for miR-580-5p. Moreover, miR-580-5p directly bound to the 3' UTR of PPARα, which decreased the production and release of C-C chemokine ligand 2 (CCL2) in HCC cells. CCL2 could activate the cyclooxygenase-2/prostaglandin E2 (COX-2/PGE2) pathway in macrophage via FoxO1 in a p38 MAPK-dependent manner. Furthermore, the Δ256 mutant of FoxO1 showed no activation effect. These results concluded that hsa_circ_0110102 acted as a sponge for miR-580-5p and inhibited CCL2 secretion into tumor microenvironment by decrease the expression of PPARα in HCC cells, then inhibited the pro-inflammatory cytokine release from macrophages by regulating the COX-2/PGE2 pathway. In conclusion, hsa_circ_0110102 served as a potential prognostic predictor or therapeutic target for HCC.
Collapse
Affiliation(s)
- Xinxing Wang
- Department of Hepatobiliary Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, Shandong, China
| | - Wei Sheng
- Department of Oncology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, Shandong, China
| | - Tao Xu
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, Shandong, China
| | - Jiawen Xu
- Department of Pathology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, Shandong, China
| | - Ruyi Gao
- Department of Hepatobiliary Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, Shandong, China
| | - Zhenhai Zhang
- Department of Hepatobiliary Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, Shandong, China
| |
Collapse
|
17
|
Shojaei-Zarghani S, Yari Khosroushahi A, Rafraf M. Oncopreventive effects of theanine and theobromine on dimethylhydrazine-induced colon cancer model. Biomed Pharmacother 2021; 134:111140. [PMID: 33360052 DOI: 10.1016/j.biopha.2020.111140] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 11/23/2020] [Accepted: 12/10/2020] [Indexed: 12/22/2022] Open
Abstract
Theanine and theobromine are abundantly present in tea and cocoa, respectively. This study was performed to assess the chemopreventive effects of these phytochemicals, alone or together, on dimethylhydrazine (DMH)-induced colon cancer. Thirty male Wistar rats were divided into five groups and subcutaneously injected with saline (negative control group) or 30 mg/kg DMH (the other groups) two times/week for 12 weeks. The negative and positive control animals were orally treated with drinking water, and the other groups were gavaged with theanine (400 mg/kg), theobromine (100 mg/kg), or their mixture for two weeks before and throughout the injection period. At the end of the study, the morphological and histopathological features, Ki-67 proliferation marker, and the expression of Akt/mTOR, JAK2/STAT3, MAPK/ERK, and TGF-β/Smad pathways were investigated. Theanine and theobromine, alone or together, reduced the number of cancerous and precancerous lesions, the volume of tumors, the Ki-67 immunostaining, and the expression of Akt/mTOR and JAK2/STAT3 oncogenic pathways. The simultaneous treatment was more effective in the down-regulation of Akt and mTOR compared to either theanine or theobromine alone. Theobromine administration also caused more inhibitory effects on the Ki-67 and Akt/mTOR expression than theanine. Besides, all dietary interventions increased the mRNA and protein expression of Smad2. In conclusion, theanine and theobromine, alone and in combination, inhibited tumorigenesis through down-regulation of the Akt/mTOR and JAK2/STAT3 pathways and an increment of the Smad2 tumor suppressor. The inhibition of the Akt/mTOR pathway was more pronounced by simultaneous treatment.
Collapse
Affiliation(s)
- Sara Shojaei-Zarghani
- Student Research Committee, Faculty of Nutrition and Food Science, Tabriz University of Medical Sciences, Tabriz, Iran; Nutrition Research Center, Department of Community Nutrition, Faculty of Nutrition and Food Science, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ahmad Yari Khosroushahi
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Medical Nanotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Maryam Rafraf
- Nutrition Research Center, Department of Community Nutrition, Faculty of Nutrition and Food Science, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
18
|
Gul HF, Ilhan N, Ilhan N, Ozercan IH, Kuloglu T. The combined effect of pomegranate extract and tangeretin on the DMBA-induced breast cancer model. J Nutr Biochem 2020; 89:108566. [PMID: 33326843 DOI: 10.1016/j.jnutbio.2020.108566] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 10/16/2020] [Accepted: 11/25/2020] [Indexed: 12/14/2022]
Abstract
The aim of this study was to investigate the protective effects of pomegranate extract and tangeretin alone or in combination in DMBA-induced rat breast cancer model. A total of 68 female rats were randomly divided into 8 groups. The first 4 groups were designed as controls for cancer and treatment groups, and the control groups were composed of only control (C), Pomegranate (P), Tangeretin (T), and Pomegranate+Tangeretin (P+T) groups. The other four groups were designed as cancer and treatment groups and were composed of DMBA (D) and DMBA+Pomegranate (D+P), DMBA+Tangeretin (D+T), DMBA+Pomegranate+Tangeretin (D+P+T) groups. Tumor markers and angiogenesis parameters were studied from plasma samples obtained from rats. Histopathological, immunohistochemical, and TUNEL analyses and expressions of proteins affecting apoptosis and cell cycle were determined in breast tissue samples. In the DMBA group, plasma CA15-3, CEA, VEGF, MMP-9, and NF-κB levels were significantly increased compared to the controls, but significant decreases were observed in these parameters except MMP-9 in the treatment groups. It was observed that p53 and Bax expressions significantly increased in both D+P and D+P+T groups compared to the DMBA group, and these findings were supported by Tunel and immunohistochemical findings. Cyclin D1 expressions were found to be significantly decreased only in the D+T group and supported by TUNEL and immunohistochemical findings. Immunohistochemical ER-α and Ki-67 immune reactivities were significantly decreased in all treatment groups compared to the DMBA group. Our results showed that combined application of pomegranate extract and tangeretin may be more beneficial in preventing breast cancer development.
Collapse
Affiliation(s)
- Huseyin Fatih Gul
- Department of Medical Biochemistry, Kafkas University, Faculty of Medicine, Kars, Turkey.
| | - Necip Ilhan
- Department of Medical Biochemistry, Firat University, Faculty of Medicine, Elazıg, Turkey
| | - Nevin Ilhan
- Department of Medical Biochemistry, Firat University, Faculty of Medicine, Elazıg, Turkey
| | | | - Tuncay Kuloglu
- Department of Histology and Embryology, Firat University, Faculty of Medicine, Elazıg, Turkey
| |
Collapse
|
19
|
Kasprzak A. Angiogenesis-Related Functions of Wnt Signaling in Colorectal Carcinogenesis. Cancers (Basel) 2020; 12:cancers12123601. [PMID: 33276489 PMCID: PMC7761462 DOI: 10.3390/cancers12123601] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 11/29/2020] [Accepted: 12/01/2020] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Angiogenesis belongs to the most clinical characteristics of colorectal cancer (CRC) and is strongly linked to the activation of Wnt/β-catenin signaling. The most prominent factors stimulating constitutive activation of this pathway, and in consequence angiogenesis, are genetic alterations (mainly mutations) concerning APC and the β-catenin encoding gene (CTNNB1), detected in a large majority of CRC patients. Wnt/β-catenin signaling is involved in the basic types of vascularization (sprouting and nonsprouting angiogenesis), vasculogenic mimicry as well as the formation of mosaic vessels. The number of known Wnt/β-catenin signaling components and other pathways interacting with Wnt signaling, regulating angiogenesis, and enabling CRC progression continuously increases. This review summarizes the current knowledge about the role of the Wnt/Fzd/β-catenin signaling pathway in the process of CRC angiogenesis, aiming to improve the understanding of the mechanisms of metastasis as well as improvements in the management of this cancer. Abstract Aberrant activation of the Wnt/Fzd/β-catenin signaling pathway is one of the major molecular mechanisms of colorectal cancer (CRC) development and progression. On the other hand, one of the most common clinical CRC characteristics include high levels of angiogenesis, which is a key event in cancer cell dissemination and distant metastasis. The canonical Wnt/β-catenin downstream signaling regulates the most important pro-angiogenic molecules including vascular endothelial growth factor (VEGF) family members, matrix metalloproteinases (MMPs), and chemokines. Furthermore, mutations of the β-catenin gene associated with nuclear localization of the protein have been mainly detected in microsatellite unstable CRC. Elevated nuclear β-catenin increases the expression of many genes involved in tumor angiogenesis. Factors regulating angiogenesis with the participation of Wnt/β-catenin signaling include different groups of biologically active molecules including Wnt pathway components (e.g., Wnt2, DKK, BCL9 proteins), and non-Wnt pathway factors (e.g., chemoattractant cytokines, enzymatic proteins, and bioactive compounds of plants). Several lines of evidence argue for the use of angiogenesis inhibition in the treatment of CRC. In the context of this paper, components of the Wnt pathway are among the most promising targets for CRC therapy. This review summarizes the current knowledge about the role of the Wnt/Fzd/β-catenin signaling pathway in the process of CRC angiogenesis, aiming to improve the understanding of the mechanisms of metastasis as well as improvements in the management of this cancer.
Collapse
Affiliation(s)
- Aldona Kasprzak
- Department of Histology and Embryology, Poznan University of Medical Sciences, Swiecicki Street 6, 60-781 Poznań, Poland
| |
Collapse
|
20
|
Aventurado CA, Billones JB, Vasquez RD, Castillo AL. In Ovo and In Silico Evaluation of the Anti-Angiogenic Potential of Syringin. Drug Des Devel Ther 2020; 14:5189-5204. [PMID: 33268982 PMCID: PMC7701684 DOI: 10.2147/dddt.s271952] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 10/08/2020] [Indexed: 01/07/2023] Open
Abstract
INTRODUCTION Cancer is considered as one of the deadliest human diseases today. Angiogenesis, the propagation of new blood vessels from pre-existing vasculature, is a critical step in the progression of cancer as it is essential in the growth and metastasis of tumors. Hence, suppression of angiogenesis is a promising approach in cancer therapy. Syringin, a phenylpropanoid glycoside with a molecular formula of C17H24O9, has been found to exhibit chemopreventive effects. However, its anti-angiogenic activity and the underlying mechanism of action are still unknown. METHODS In this work, in ovo chorioallantoic membrane (CAM) assay has been conducted to evaluate the effect of syringin on neovascularization. Additionally, reverse molecular docking studies have been performed in order to identify the probable enzyme targets in the angiogenesis pathway. RESULTS Treatment with syringin showed significant dose-dependent inhibition of blood vessel length and junctions in the CAM of duck eggs; the anti-angiogenic activity of syringin at 100 µM and 200 µM is comparable with 200 µM of the positive control celecoxib. The results of reverse docking studies indicate that syringin binds the strongest to dihydrofolate reductase (DHFR) and, to some extent, with transforming growth factor-beta receptor type 1 (TGF-βR1), vascular endothelial growth factor receptor 2 (VEGFR2), and matrix metalloproteinase-2 (MMP-2). Furthermore, ADMET models revealed that syringin potentially possesses excellent pharmacokinetic and toxicity profiles. CONCLUSION This study demonstrates the potential of syringin as an anti-angiogenic agent and elicits further investigations to establish its application in cancer suppression.
Collapse
Affiliation(s)
| | - Junie B Billones
- Department of Physical Sciences and Mathematics, College of Arts and Sciences, University of the Philippines Manila, Manila, Philippines
| | - Ross D Vasquez
- The Graduate School, University of Santo Tomas, Manila1015, Philippines
- Faculty of Pharmacy, University of Santo Tomas, Manila1015, Philippines
- Research Center for the Natural and Applied Sciences, University of Santo Tomas, Manila1015, Philippines
| | - Agnes L Castillo
- The Graduate School, University of Santo Tomas, Manila1015, Philippines
- Faculty of Pharmacy, University of Santo Tomas, Manila1015, Philippines
- Research Center for the Natural and Applied Sciences, University of Santo Tomas, Manila1015, Philippines
| |
Collapse
|
21
|
Kim J, Noh MH, Hur DY, Kim B, Kim YS, Lee HK. Celecoxib upregulates ULBP-1 expression in lung cancer cells via the JNK/PI3K signaling pathway and increases susceptibility to natural killer cell cytotoxicity. Oncol Lett 2020; 20:279. [PMID: 33014157 PMCID: PMC7520723 DOI: 10.3892/ol.2020.12142] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Accepted: 09/08/2020] [Indexed: 01/22/2023] Open
Abstract
Lung cancer has the highest cancer mortality rate in the world, and effective therapies are still required. Cyclooxygenase-2 (COX-2) is highly expressed in numerous types of cancer, and is therefore considered a possible target of cancer treatment. Celecoxib, a selective COX-2 inhibitor, has binding pockets that interact with COX-2 and disrupt its enzymatic activities. In addition, celecoxib is able to affect cellular functions in a COX-2-independent manner. The present study aimed to investigate if celecoxib affected natural killer (NK) cell receptors and susceptibility to NK cell toxicity. For this purpose, PCR, immunoblotting, flow cytometry analysis and NK cell cytotoxicity assays were performed. The present study revealed that sublethal concentrations of celecoxib increased the expression levels of UL16-binding protein 1 (ULBP-1), a natural-killer group 2 member D (NKG2D) ligand, in lung cancer A549 and H460 cell lines. ULBP-1 mRNA and protein expression was induced in a dose- and time-dependent manner after celecoxib treatment. Expression levels of other NKG2D ligands, such as ULBP-2, ULBP-3, MHC class I-related chain A (MICA) and MICB did not change considerably compared to ULBP-1 in response to celecoxib treatment. Fluorescence microscopic images revealed abundant ULBP-1 in the cytoplasm after celecoxib treatment. Both JNK and PI3K may be involved in the induction of ULBP-1 expression after celecoxib treatment in A549 and H460 cells. In a NK cytotoxicity assay, celecoxib increased the sensitivity to NK cell-mediated cytotoxicity via interaction with ULBP-1 in lung cancer cells. Overall, the present results demonstrated that celecoxib treatment induced ULBP-1 expression in lung cancer cells, thereby increasing their susceptibility to NK cell cytotoxicity. These results suggest that the effects of conventional anticancer therapy may potentially be enhanced by using celecoxib, which targets COX-2, to enhance the sensitivity of lung cancer cells to NK cell-mediated cytotoxicity.
Collapse
Affiliation(s)
- Jayoung Kim
- Department of Medical Science, Pusan National University School of Medicine, Yangsan, South Gyeongsang 50612, Republic of Korea
| | - Min Hye Noh
- Department of Anatomy, Inje University College of Medicine, Busan 47396, Republic of Korea
| | - Dae Young Hur
- Department of Anatomy, Inje University College of Medicine, Busan 47396, Republic of Korea
| | - Bomi Kim
- Department of Pathology, Inje University Haewoondae Paik Hospital, Busan 48108, Republic of Korea
| | - Yeong Seok Kim
- Department of Anatomy, Inje University College of Medicine, Busan 47396, Republic of Korea
| | - Hyun-Kyung Lee
- Division of Pulmonology, Department of Internal Medicine, Inje University Pusan Paik Hospital, Busan 47396, Republic of Korea
| |
Collapse
|
22
|
Celecoxib, a selective COX-2 inhibitor, markedly reduced the severity of tamoxifen-induced adenomyosis in a murine model. Exp Ther Med 2020; 19:3289-3299. [PMID: 32266025 PMCID: PMC7132242 DOI: 10.3892/etm.2020.8580] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 12/05/2019] [Indexed: 12/17/2022] Open
Abstract
The aim of the present study was to evaluate the effects of the selective cyclooxygenase (COX)-2 inhibitor celecoxib on the development of uterine adenomyosis in mice. ICR neonatal mice were first exposed to tamoxifen to establish a mouse model of adenomyosis. Following 60 days of celecoxib treatment, pathological formation of adenomyosis lesions and the depth of myometrial infiltration were evaluated using hematoxylin and eosin staining. To examine thermal pain modulation in mice, a hotplate test was conducted every 15 days from postnatal day 30 onwards. Immunohistochemistry was performed to assess the expression of aromatase P450, N-cadherin, E-cadherin, COX-2 and cluster of differentiation 31, whereas the levels of estrogen were analyzed in uterine tissue homogenates using ELISA. Masson trichrome staining was performed to assess the extent of fibrosis in the uterus. Celecoxib treatment significantly inhibited the depth of infiltration into the myometrium, resulting in significantly reduced disease severity. Treatment with high doses of celecoxib significantly prolonged thermal response latency. Following celecoxib treatment, the expression of E-cadherin was significantly increased whereas the expression of N-cadherin was significantly decreased. Concomitantly, the extent of fibrosis was also reduced following celecoxib treatment. Uterine tissue homogenates isolated from mice treated with both high and low doses of celecoxib exhibited lower concentrations of estrogen and decreased expression of aromatase P450. These observations suggest that celecoxib reduces adenomyosis severity by suppressing estrogen production in the uterus, reversing epithelial-mesenchymal transition and relieving fibrosis. Taken together, the results of the present study support the potential use of celecoxib, a selective COX-2 inhibitor, for the treatment of adenomyosis.
Collapse
|
23
|
COX-2 Signaling in the Tumor Microenvironment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1277:87-104. [PMID: 33119867 DOI: 10.1007/978-3-030-50224-9_6] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Tumorigenesis is a multistep, complicated process, and many studies have been completed over the last few decades to elucidate this process. Increasingly, many studies have shifted focus toward the critical role of the tumor microenvironment (TME), which consists of cellular players, cell-cell communications, and extracellular matrix (ECM). In the TME, cyclooxygenase-2 (COX-2) has been found to be a key molecule mediating the microenvironment changes. COX-2 is an inducible form of the enzyme that converts arachidonic acid into the signal transduction molecules (thromboxanes and prostaglandins). COX-2 is frequently expressed in many types of cancers and has been closely linked to its occurrence, progression, and prognosis. For example, COX-2 has been shown to (1) regulate tumor cell growth, (2) promote tissue invasion and metastasis, (3) inhibit apoptosis, (4) suppress antitumor immunity, and (5) promote sustainable angiogenesis. In this chapter, we summarize recent advances of studies that have evaluated COX-2 signaling in TME.
Collapse
|
24
|
Irani YD, Pulford E, Mortimer L, Irani S, Butler L, Klebe S, Williams KA. Sex differences in corneal neovascularization in response to superficial corneal cautery in the rat. PLoS One 2019; 14:e0221566. [PMID: 31479468 PMCID: PMC6719872 DOI: 10.1371/journal.pone.0221566] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Accepted: 08/10/2019] [Indexed: 12/23/2022] Open
Abstract
Sex-based differences in susceptibility have been reported for a number of neovascular ocular diseases. We quantified corneal neovascularization, induced by superficial silver nitrate cautery, in male and female inbred albino Sprague-Dawley, inbred albino Fischer 344, outbred pigmented Hooded Wistar and inbred pigmented Dark Agouti rats of a range of ages. Corneal neovascular area was quantified on haematoxylin-stained corneal flatmounts by image analysis. Pro-and anti-angiogenic gene expression was measured early in the neovascular response by quantitative real-time polymerase chain reaction. Androgen and estrogen receptor expression was assessed by immunohistochemistry. Male rats from all strains, with or without ocular pigmentation, exhibited significantly greater corneal neovascular area than females: Sprague-Dawley males 43±12% (n = 8), females 25±5% (n = 12), p = 0.001; Fischer 344 males 38±10% (n = 12) females 27±8% (n = 8) p = 0.043; Hooded Wistar males 32±6% (n = 8) females 22±5% (n = 12) p = 0.002; Dark Agouti males 37±11% (n = 9) females 26±7% (n = 9) p = 0.015. Corneal vascular endothelial cells expressed neither androgen nor estrogen receptor. The expression in cornea post-cautery of Cox-2, Vegf-a and Vegf-r2 was significantly higher in males compared with females and Vegf-r1 was significantly lower in the cornea of males compared to females, p<0.001 for each comparison. These data suggest that male corneas are primed for angiogenesis through a signalling nexus involving Cox-2, Vegf-a, and Vegf receptors 1 and 2. Our findings re-enforce that pre-clinical animal models of human diseases should account for sex-based differences in their design and highlight the need for well characterized and reproducible pre-clinical studies that include both male and female animals.
Collapse
Affiliation(s)
- Yazad D. Irani
- Discipline of Ophthalmology, College of Medicine and Public Health, Flinders University, Adelaide, Australia
| | - Emily Pulford
- Discipline of Anatomical Pathology, College of Medicine and Public Health, Flinders University, Adelaide, Australia
| | - Lauren Mortimer
- Discipline of Ophthalmology, College of Medicine and Public Health, Flinders University, Adelaide, Australia
- Discipline of Anatomical Pathology, College of Medicine and Public Health, Flinders University, Adelaide, Australia
| | - Swati Irani
- Freemasons Foundation Centre for Men's Health, Adelaide Medical School, University of Adelaide, Adelaide, Australia
- South Australian Health and Medical Research Institute, Adelaide, Australia
| | - Lisa Butler
- Freemasons Foundation Centre for Men's Health, Adelaide Medical School, University of Adelaide, Adelaide, Australia
- South Australian Health and Medical Research Institute, Adelaide, Australia
| | - Sonja Klebe
- Discipline of Anatomical Pathology, College of Medicine and Public Health, Flinders University, Adelaide, Australia
| | - Keryn A. Williams
- Discipline of Ophthalmology, College of Medicine and Public Health, Flinders University, Adelaide, Australia
| |
Collapse
|
25
|
Resveratrol Action on Lipid Metabolism in Cancer. Int J Mol Sci 2019; 20:ijms20112704. [PMID: 31159437 PMCID: PMC6601040 DOI: 10.3390/ijms20112704] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 05/27/2019] [Accepted: 05/28/2019] [Indexed: 12/12/2022] Open
Abstract
Cancer diseases have the leading position in human mortality nowadays. The age of oncologic patients is still decreasing, and the entire scientific society is eager for new ways to fight against cancer. One of the most discussed issues is prevention by means of natural substances. Resveratrol is a naturally occurring plant polyphenol with proven antioxidant, anti-inflammatory, and anticancer effects. Tumor cells display specific changes in the metabolism of various lipids. Resveratrol alters lipid metabolism in cancer, thereby affecting storage of energy, cell signaling, proliferation, progression, and invasiveness of cancer cells. At the whole organism level, it contributes to the optimal metabolism extent with respect to the demands of the organism. Thus, resveratrol could be used as a preventive and anticancer agent. In this review, we focus on some of the plethora of lipid pathways and signal molecules which are affected by resveratrol during carcinogenesis.
Collapse
|
26
|
Fu X, Zhang H, Chen Z, Yang Z, Shi D, Liu T, Chen W, Yao F, Su X, Deng W, Chen M, Yang A. TFAP2B overexpression contributes to tumor growth and progression of thyroid cancer through the COX-2 signaling pathway. Cell Death Dis 2019; 10:397. [PMID: 31113934 PMCID: PMC6529436 DOI: 10.1038/s41419-019-1600-7] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 04/01/2019] [Accepted: 04/08/2019] [Indexed: 12/13/2022]
Abstract
Thyroid cancer is commonly seen in the clinic with a rapidly increasing incidence globally. COX-2 overexpression correlates with the pathologic type of thyroid carcinoma, and it has been suggested that COX-2 overexpression is associated with a poor prognosis. However, little is known about its upstream regulatory mechanism. Bioinformatics suggested that transcription factor AP-2 beta (TFAP2B) might specifically bind to the COX-2 promoter, which was confirmed by biotin-labeled COX-2 promoter pulldown and luciferase reporter assays. We performed western blot and immunohistochemical staining to detect the expression of TFAP2B/COX-2 in thyroid cancer tissues (T) and the matched adjacent noncarcinoma tissues (ANT), and investigated the relationship between TFAP2B/COX-2 expression and clinical pathological factors in thyroid cancer patients. Afterward, MTS, colony formation, cell-apoptosis assay, transwell-invasion and scratch assays were performed to examine the proliferation, apoptosis, invasion, and migration of thyroid cancer cells with TFAP2B knocked down or overexpressed. The mouse xenograft experiment was performed to study in vivo the proliferation of thyroid cancer cells with TFAP2B knocked down or overexpressed. We found that TFAP2B bound to the promoter of COX-2 to activate its expression. Western blot and immunohistochemistry showed that TFAP2B/COX-2 was highly expressed in thyroid cancer, and high TFAP2B and COX-2 expression was associated with aggressive clinicopathological features in thyroid cancer. TFAP2B mediated thyroid cancer cell proliferation, apoptosis, invasion, and migration via the COX-2 signaling pathway in vitro and in vivo. TFAP2B bound to the promoter of COX-2 to activate its expression, indicating that TFAP2B is a critical regulatory molecule in the COX-2 signaling pathway that promoted tumor progression in thyroid cancer.
Collapse
Affiliation(s)
- Xiaoyan Fu
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center of Cancer Medicine, Guangzhou, Guangdong, China
| | - Huayong Zhang
- Department of Thyroid and Breast Surgery, the Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, China
| | - Zhipeng Chen
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center of Cancer Medicine, Guangzhou, Guangdong, China
| | - Zhongyuan Yang
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center of Cancer Medicine, Guangzhou, Guangdong, China
| | - Dingbo Shi
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center of Cancer Medicine, Guangzhou, Guangdong, China
| | - Tianrun Liu
- Department of Otorhinolaryngology Head and Neck Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Weichao Chen
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center of Cancer Medicine, Guangzhou, Guangdong, China
| | - Fan Yao
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center of Cancer Medicine, Guangzhou, Guangdong, China
| | - Xuan Su
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center of Cancer Medicine, Guangzhou, Guangdong, China
| | - Wuguo Deng
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center of Cancer Medicine, Guangzhou, Guangdong, China.
| | - Miao Chen
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center of Cancer Medicine, Guangzhou, Guangdong, China.
| | - Ankui Yang
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center of Cancer Medicine, Guangzhou, Guangdong, China.
| |
Collapse
|
27
|
Li Y, Kakkar R, Wang J. In vivo and in vitro Approach to Anti-arthritic and Anti-inflammatory Effect of Crocetin by Alteration of Nuclear Factor-E2-Related Factor 2/hem Oxygenase (HO)-1 and NF-κB Expression. Front Pharmacol 2018; 9:1341. [PMID: 30618728 PMCID: PMC6299880 DOI: 10.3389/fphar.2018.01341] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2018] [Accepted: 10/31/2018] [Indexed: 01/15/2023] Open
Abstract
Crocetin (apo carotenoid dicarboxylic acid) is a common constituent of saffron. Its importance is well documented in Chinese medicine. Some studies have reported the inhibitory effect on inflammation in rats. The aim of the current experimental investigation to scrutinize the anti-inflammatory effect of Crocetin using the lipo polysaccharide (LPS) induced mouse macrophages (RAW 264.7) in vitro and complete Freund’s adjuvant-induced arthritis model and to explore in vivo possible mechanism of action. RAW 264.7 macrophages were used for estimation of the effect of crocetin on the cyclooxygenase (COX-2), nitric oxide (NO)production, anti-inflammatory and along with pro-inflammatory cytokines such as tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β), interleukin-6 (IL-6), and interleukin-10 (IL-10). Single intraperitoneal injection of complete freund’s adjuvant (CFA) was used to induce arthritis. The rats were divided into different group and received the oral administration of crocetin in a dose-dependent manner with indomethacin till 28 days. The paw edema and body weight was estimated at regular interval of time. The biochemical parameters, hematological and pro-inflammatory cytokines such as tumor necrosis factor receptor 1 (TNF-R1), IL-6, and IL-1β, Vascular endothelial growth factor (VEGF); heme oxygenase-1/nuclear factor erythroid 2–related factor 2 (HO-1/Nrf-2) expression were estimated at end of the experimental study. Crocetin inhibited the COX-2 catalyzed prostaglandin (PGE2) and inducible nitric oxide synthase catalyzed NO production on RAW 264.7. The paw edema and body weight was significantly (P < 0.001) modulated by the Crocetin in a dose-dependent manner. Crocetin treatment increased the level of red blood cells (RBC), hemoglobin (Hb) and decreased level of white blood cells (WBC), erythrocyte sedimentation rate (ESR), alkaline phosphatase (ALP), serum glutamic pyruvic transaminase (SGPT), and serum glutamic-oxaloacetic transaminase (SGOT) parameters, with reduction of TNF-α, IL-6, and IL-1β.The protective effect of crocetin was substantiated with a reduction in expression of IL-6, IL-1β, VEGF, and TNF-R1, respectively. Crocetin also increased the HO-1/Nrf-2 and decreased the nuclear factor kappa-B (NF-κB) mRNA, protein expression. On the basis of the result, we can conclude that the reduction of HO-1/Nrf-2 expression, as well as inflammatory mediators, may be involved in the protective effect of Crocetin in the CFA model.
Collapse
Affiliation(s)
- Yi Li
- Department of Joint Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
| | - Rajat Kakkar
- Chandrasheker College of Pharmacy, Allahabad, India
| | - Jian Wang
- Department of Joint Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
| |
Collapse
|
28
|
Hashemi Goradel N, Najafi M, Salehi E, Farhood B, Mortezaee K. Cyclooxygenase-2 in cancer: A review. J Cell Physiol 2018; 234:5683-5699. [PMID: 30341914 DOI: 10.1002/jcp.27411] [Citation(s) in RCA: 505] [Impact Index Per Article: 72.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2018] [Accepted: 08/22/2018] [Indexed: 12/17/2022]
Abstract
Cyclooxygenase-2 (COX-2) is frequently expressed in many types of cancers exerting a pleiotropic and multifaceted role in genesis or promotion of carcinogenesis and cancer cell resistance to chemo- and radiotherapy. COX-2 is released by cancer-associated fibroblasts (CAFs), macrophage type 2 (M2) cells, and cancer cells to the tumor microenvironment (TME). COX-2 induces cancer stem cell (CSC)-like activity, and promotes apoptotic resistance, proliferation, angiogenesis, inflammation, invasion, and metastasis of cancer cells. COX-2 mediated hypoxia within the TME along with its positive interactions with YAP1 and antiapoptotic mediators are all in favor of cancer cell resistance to chemotherapeutic drugs. COX-2 exerts most of the functions through its metabolite prostaglandin E2. In some and limited situations, COX-2 may act as an antitumor enzyme. Multiple signals are contributed to the functions of COX-2 on cancer cells or its regulation. Members of mitogen-activated protein kinase (MAPK) family, epidermal growth factor receptor (EGFR), and nuclear factor-κβ are main upstream modulators for COX-2 in cancer cells. COX-2 also has interactions with a number of hormones within the body. Inhibition of COX-2 provides a high possibility to exert therapeutic outcomes in cancer. Administration of COX-2 inhibitors in a preoperative setting could reduce the risk of metastasis in cancer patients. COX-2 inhibition also sensitizes cancer cells to treatments like radio- and chemotherapy. Chemotherapeutic agents adversely induce COX-2 activity. Therefore, choosing an appropriate chemotherapy drugs along with adjustment of the type and does for COX-2 inhibitors based on the type of cancer would be an effective adjuvant strategy for targeting cancer.
Collapse
Affiliation(s)
- Nasser Hashemi Goradel
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Masoud Najafi
- Radiology and Nuclear Medicine Department, School of Paramedical Sciences, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Eniseh Salehi
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Bagher Farhood
- Departments of Medical Physics and Radiology, Faculty of Paramedical Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Keywan Mortezaee
- Department of Anatomy, School of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran
| |
Collapse
|
29
|
Peng Y, Wang Y, Tang N, Sun D, Lan Y, Yu Z, Zhao X, Feng L, Zhang B, Jin L, Yu F, Ma X, Lv C. Andrographolide inhibits breast cancer through suppressing COX-2 expression and angiogenesis via inactivation of p300 signaling and VEGF pathway. J Exp Clin Cancer Res 2018; 37:248. [PMID: 30314513 PMCID: PMC6186120 DOI: 10.1186/s13046-018-0926-9] [Citation(s) in RCA: 113] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Accepted: 10/02/2018] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Andrographolide (Andro), a diterpenoid lactone, has been used for treatment of various cancers with less adverse effects. However, the underlying mechanisms regarding its anti-tumor mechanism still remain unclear. METHODS Cell viability and proliferation were measured by CCK8 and CFSE dilution assay. The localization of p50/p65 or cytochrome c was determined using confocal immunofluorescence. Streptavidin-agarose pulldown or ChIP assays were used to detect the binding of multiple transactivators to COX-2 promoter. The promoter activity was examined by a dual-Luciferase reporter assay. The functions of Andro on COX-2-mediated angiogenesis were also investigated using human HUVEC cells through tube formation and spheroids sprouting assay. The in vivo anti-tumor efficacy of Andro was analyzed in xenografts nude mice. RESULTS The results indicated that Andro could significantly inhibit the proliferation of human breast cancers, and suppress COX-2 expression at both protein and mRNA levels. Furthermore, Andro could dose-dependently inhibit COX-2-mediated angiogenesis in human endothelial cells. We have also found that Andro significantly promoted the activation of cytochrome c and activated caspase-dependent apoptotic signaling pathway. Our further explorations demonstrated that Andro inhibited the binding of the transactivators CREB2, C-Fos and NF-κB and blocked the recruitment of coactivator p300 to COX-2 promoter. Moreover, Andro could effectively inhibit the activity of p300 histone acetyltransferase (HAT), thereby attenuating the p300-mediated acetylation of NF-κB. Besides, Andro could also dramatically inhibit the migration, invasion and tubulogenesis of HUVECs in vitro. In addition, Andro also exhibited effective anti-tumor efficacy as well as angiogenesis inhibition in vivo. CONCLUSION In current study, we explore the potential effects of Andro in suppressing breast cancer growth and tumor angiogenesis, as well as the precise mechanisms. This work demonstrated the potential anti-cancer effects of Andro, indicating that Andro could inhibit COX-2 expression through attenuating p300 HAT activity and suppress angiogenesis via VEGF pathway, and thereby could be developed as an antitumor agent for the treatment of breast cancer.
Collapse
Affiliation(s)
- Yulin Peng
- Institute of Integrative Medicine, College of Pharmacy, College of Basic Medical Science, Dalian Medical University, Dalian, 116044 China
| | - Yan Wang
- Institute of Integrative Medicine, College of Pharmacy, College of Basic Medical Science, Dalian Medical University, Dalian, 116044 China
| | - Ning Tang
- Institute of Integrative Medicine, College of Pharmacy, College of Basic Medical Science, Dalian Medical University, Dalian, 116044 China
- Department of Integrative Medicine, Liaoning University of Traditional Chinese Medicine Xinglin College, Shenyang, 110167 China
| | - Dongdong Sun
- Institute of Integrative Medicine, College of Pharmacy, College of Basic Medical Science, Dalian Medical University, Dalian, 116044 China
| | - Yulong Lan
- Institute of Integrative Medicine, College of Pharmacy, College of Basic Medical Science, Dalian Medical University, Dalian, 116044 China
| | - Zhenlong Yu
- Institute of Integrative Medicine, College of Pharmacy, College of Basic Medical Science, Dalian Medical University, Dalian, 116044 China
- Emergency Department, The Second Affiliated Hospital of Hainan Medical University, Haikou, 571199 China
| | - Xinyu Zhao
- Institute of Integrative Medicine, College of Pharmacy, College of Basic Medical Science, Dalian Medical University, Dalian, 116044 China
| | - Lei Feng
- Institute of Integrative Medicine, College of Pharmacy, College of Basic Medical Science, Dalian Medical University, Dalian, 116044 China
- Emergency Department, The Second Affiliated Hospital of Hainan Medical University, Haikou, 571199 China
| | - Baojing Zhang
- Institute of Integrative Medicine, College of Pharmacy, College of Basic Medical Science, Dalian Medical University, Dalian, 116044 China
| | - Lingling Jin
- Institute of Integrative Medicine, College of Pharmacy, College of Basic Medical Science, Dalian Medical University, Dalian, 116044 China
| | - Fabiao Yu
- Emergency Department, The Second Affiliated Hospital of Hainan Medical University, Haikou, 571199 China
| | - Xiaochi Ma
- Institute of Integrative Medicine, College of Pharmacy, College of Basic Medical Science, Dalian Medical University, Dalian, 116044 China
- Emergency Department, The Second Affiliated Hospital of Hainan Medical University, Haikou, 571199 China
| | - Chuanzhu Lv
- Emergency Department, The Second Affiliated Hospital of Hainan Medical University, Haikou, 571199 China
| |
Collapse
|
30
|
Zhang J, He J, Zhang L. The down-regulation of microRNA-137 contributes to the up-regulation of retinoblastoma cell proliferation and invasion by regulating COX-2/PGE2 signaling. Biomed Pharmacother 2018; 106:35-42. [PMID: 29945115 DOI: 10.1016/j.biopha.2018.06.099] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Revised: 06/13/2018] [Accepted: 06/13/2018] [Indexed: 02/07/2023] Open
Abstract
MicroRNA-137 (miR-137) plays an important role in the development and progression of many types of human cancers; however, the role of miR-137 in retinoblastoma (RB) remains unclear. In this study, we aimed to investigate the functional significance and molecular mechanisms of miR-137 in RB. We reported that miR-137 was frequently down-regulated in RB tissues and cell lines. The overexpression of miR-137 inhibited RB cell proliferation and invasion, while the suppression of miR-137 promoted RB cell proliferation and invasion. Bioinformatic analysis predicted that cyclooxygenase-2 (COX-2) was a potential target gene of miR-137, which was validated by a dual-luciferase reporter assay. Moreover, our results showed that miR-137 negatively regulated the expression of COX-2 and the production of prostaglandin E2 (PGE2) in RB cells. The knockdown of COX-2 suppressed the proliferation and invasion of RB cells as well as the production of PGE2. The overexpression of COX-2 significantly reversed the inhibitory effect of miR-137 overexpression on RB cell proliferation and invasion. Taken together, these results suggest that miR-137 suppresses the proliferation and invasion of RB cells by targeting COX-2/PGE2. Our study reveals a tumor suppressive role of miR-137 in the progression of RB and suggests miR-137 as a potentially effective therapeutic target for the treatment of RB.
Collapse
Affiliation(s)
- Jian Zhang
- Department of Ophthalmology, Shaanxi Provincial People's Hospital, Xi'an, 710068, China
| | - Jing He
- Department of Obstetrics, The First Affiliated Hospital of Xi'an Medical University, Xi'an, 710077, China.
| | - Le Zhang
- Department of Ophthalmology, Shaanxi Provincial People's Hospital, Xi'an, 710068, China
| |
Collapse
|