1
|
Shafi FAA, Imeer ATA, Nassrullah HAA, Naeemah AM. The impact of extracellular glucose concentrations on antioxidant capacity, viability, and microRNA expression in TM4 Sertoli cells. Reprod Biol 2025; 25:101015. [PMID: 40163939 DOI: 10.1016/j.repbio.2025.101015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Revised: 03/09/2025] [Accepted: 03/24/2025] [Indexed: 04/02/2025]
Abstract
This study investigates the impact of extracellular glucose concentrations on antioxidant capacity, viability, and microRNA (miR) expression in TM4 Sertoli cell lines. TM4 cells were cultured in high-glucose (115 µm) and low-glucose (<505 µm) conditions to simulate hyperglycemia and glucose starvation, respectively. The study measured total antioxidant capacity (TAC), malondialdehyde (MDA), total oxidant status (TOS), glutathione (GSH), glutathione disulfide (GSSG), NADP/NADPH, glutathione peroxidase (GPX), and glutathione reductase (GR) levels. MiR-17, miR-34, miR-106a, and miR-200a expression levels were assessed. Cell viability and apoptosis were evaluated using MTT assay and acridine-orange staining. Results indicated that high glucose reduced miR-17 expression while low glucose increased it. Both glucose conditions elevated miR-34, miR-106a, and miR-200a expressions. TAC levels decreased, while TOS and MDA levels increased significantly under both conditions. High glucose had no significant effect on GPX and GR levels, whereas low glucose decreased them. Both conditions led to reduced GSH levels, increased GSSG levels, and altered NADP/NADPH ratio. Increased apoptosis and decreased cell viability were observed under both glucose conditions. These findings suggest that extracellular glucose levels significantly dysregulate miRNA expression, antioxidant capacities, and redox buffer systems in TM4 cells. High glucose conditions suppress miR-17 expression, increase miR-34 and miR-106a levels, and induce reductive buffer imbalance. Conversely, low glucose conditions trigger compensatory mechanisms via increased miR-17 expression to enhance antioxidant status while reducing GPX and GR levels. These results provide insights into the molecular responses of Sertoli cells under varying glucose environments, highlighting potential therapeutic pathways for conditions like diabetes and metabolic dysfunctions.
Collapse
Affiliation(s)
- Farha A Ali Shafi
- Department of Biology College of Science Mustansiriyah University, Baghdad, Iraq.
| | | | | | - Ali Mutashar Naeemah
- Department of Histology and Embryology, Faculty of Veterinary Medicine, Urmia University, Urmia, Iran
| |
Collapse
|
2
|
Saadh MJ, Ahmed HH, Chandra M, Al-Hussainy AF, Hamid JA, Mishra A, Taher WM, Alwan M, Jawad MJ, Al-Nuaimi AMA, Alsaikhan F, Farhood B, Akhavan-Sigari R. Therapeutic effects of quercetin in oral cancer therapy: a systematic review of preclinical evidence focused on oxidative damage, apoptosis and anti-metastasis. Cancer Cell Int 2025; 25:66. [PMID: 39994659 PMCID: PMC11854426 DOI: 10.1186/s12935-025-03694-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 02/13/2025] [Indexed: 02/26/2025] Open
Abstract
OBJECTIVE Oral malignancies are among the common head and neck cancers. Various therapeutic modalities are used for targeting oral cancers. It was shown that quercetin (a flavonoid) has an anti-cancer effect on different cancers. In the current study, the anti-cancer potentials of quercetin against oral cancer cells were summarized. METHODS The current systematic review was conducted in accordance with the PRISMA guideline for the identification of relevant studies in various electronic databases up to April 2023. After reviewing and screening 193 articles, 18 were chosen for this study based on our inclusion and exclusion criteria. RESULTS It was shown that quercetin significantly reduced cancer cell proliferation, cell viability, tumor volume, invasion, metastasis and migration. This anti-cancer agent induced oxidative stress and apoptosis in the cancer cells. Quercetin treatment could also induce some biochemical alterations in the cancer cells. CONCLUSION According to the results, it can be mentioned that quercetin administration has an anti-cancer effect against oral cancer cells. This agent exerts its anticancer effects via reduced cell viability and different mechanisms, including induce oxidative damage, apoptosis, and reduced invasion and metastasis. However, suggesting the use of quercetin as a therapeutic agent of oral cancer patients requires further clinical studies due to its poor absorption rates, and the exact molecular mechanisms are still not well understood.
Collapse
Affiliation(s)
- Mohamed J Saadh
- Faculty of Pharmacy, Middle East University, Amman, 11831, Jordan
| | | | - Muktesh Chandra
- Marwadi University Research Center, Department of Bioinformatics, Faculty of Engineering and Technology, Marwadi University, Rajkot, 360003, Gujarat, India
| | | | | | - Anurag Mishra
- Department of Pharmaceutical Chemistry, NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, India
| | - Waam Mohammed Taher
- College of Nursing, National University of Science and Technology, Dhi Qar, Iraq
| | - Mariem Alwan
- Pharmacy College, Al-Farahidi University, Baghdad, Iraq
| | | | | | - Fahad Alsaikhan
- College of Pharmacy, Prince Sattam Bin Abdulaziz University, Alkharj, Saudi Arabia
- School of Pharmacy, Ibn Sina National College for Medical Studies, Jeddah, Saudi Arabia
| | - Bagher Farhood
- Department of Medical Physics and Radiology, Faculty of Paramedical Sciences, Kashan University of Medical Sciences, Kashan, Iran.
| | - Reza Akhavan-Sigari
- Dr. Schneiderhan GmbH and ISAR Klinikum, Munich, Germany
- Department of Health Care Management and Clinical Research, Collegium Humanum Warsaw Management University Warsaw, Warszawa, Poland
| |
Collapse
|
3
|
Padmanaban D, Rajkumar P, Vijayakumar M, Jayaprakash V, Sekar N, Valayapathi R, Azhagudurai A, Chinnasamy A. Exploring sulphated polysaccharides from marine bivalves: Unveiling potent anti-gastric cancer activity through cell cycle arrest and apoptosis. JOURNAL OF ASIAN NATURAL PRODUCTS RESEARCH 2025:1-22. [PMID: 39985784 DOI: 10.1080/10286020.2025.2468319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 02/11/2025] [Accepted: 02/13/2025] [Indexed: 02/24/2025]
Abstract
This study investigates the extraction of partially purified sulphated polysaccharides from marine bivalves through enzymatic digestion followed by sequential deproteinization and evaluates their anti-gastric cancer activity. Chemical analysis confirmed high uronic acid and sulfate content, with FT-IR validating characteristic functional groups. Donax variabilis polysaccharide demonstrated significant, dose-dependent anti-proliferative effects against AGS cells. Mechanistic studies revealed morphological changes, ROS generation, LDH release, and mitochondrial membrane disruption, leading to apoptosis. Flow cytometry confirmed G0/G1 phase arrest. These findings suggest that D. variabilis sulphated polysaccharides exhibit potent anti-gastric cancer properties, likely through oxidative stress and mitochondrial dysfunction, warranting further investigation for therapeutic applications.
Collapse
Affiliation(s)
- Deepakrajasekar Padmanaban
- Department of Zoology, University of Madras, Guindy Campus, Chennai, Tamil Nadu600 025, India
- Crustacean Culture Division, ICAR-CIBA, 75, Santhome High Road, M. R. C. Nagar, R. A. Puram, Chennai, Tamil Nadu600 028, India
| | - Pavithra Rajkumar
- Department of Zoology, University of Madras, Guindy Campus, Chennai, Tamil Nadu600 025, India
- Department of Biological Sciences, SRM AP University, Mangalagiri Mandal, Guntur District, Andhra Pradesh522502, India
| | - Mahalakshmi Vijayakumar
- Department of Zoology, University of Madras, Guindy Campus, Chennai, Tamil Nadu600 025, India
| | - Vennila Jayaprakash
- Department of Zoology, University of Madras, Guindy Campus, Chennai, Tamil Nadu600 025, India
| | - Niranjni Sekar
- Department of Zoology, University of Madras, Guindy Campus, Chennai, Tamil Nadu600 025, India
| | - Rajasekar Valayapathi
- Department of Zoology, University of Madras, Guindy Campus, Chennai, Tamil Nadu600 025, India
| | - Aarthi Azhagudurai
- Department of Zoology, University of Madras, Guindy Campus, Chennai, Tamil Nadu600 025, India
| | - Arulvasu Chinnasamy
- Department of Zoology, University of Madras, Guindy Campus, Chennai, Tamil Nadu600 025, India
| |
Collapse
|
4
|
Naponelli V, Piscazzi A, Mangieri D. Cellular and Molecular Mechanisms Modulated by Genistein in Cancer. Int J Mol Sci 2025; 26:1114. [PMID: 39940882 PMCID: PMC11818640 DOI: 10.3390/ijms26031114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 01/21/2025] [Accepted: 01/23/2025] [Indexed: 02/16/2025] Open
Abstract
Genistein (4',5,7-trihydroxyisoflavone) is a phytoestrogen belonging to a subclass of natural flavonoids that exhibits a wide range of pharmacological functions, including antioxidant and anti-inflammatory properties. These characteristics make genistein a valuable phytochemical compound for the prevention and/or treatment of cancer. Genistein effectively inhibits tumor growth and dissemination by modulating key cellular mechanisms. This includes the suppression of angiogenesis, the inhibition of epithelial-mesenchymal transition, and the regulation of cancer stem cell proliferation. These effects are mediated through pivotal signaling pathways such as JAK/STAT, PI3K/Akt/mTOR, MAPK/ERK, NF-κB, and Wnt/β-catenin. Moreover, genistein interferes with the function of specific cyclin/CDK complexes and modulates the activation of Bcl-2/Bax and caspases, playing a critical role in halting tumor cell division and promoting apoptosis. The aim of this review is to discuss in detail the key cellular and molecular mechanisms underlying the pleiotropic anticancer effects of this flavonoid.
Collapse
Affiliation(s)
- Valeria Naponelli
- Department of Medicine and Surgery, University of Parma, Plesso Biotecnologico Integrato, Via Volturno 39, 43126 Parma, Italy
| | - Annamaria Piscazzi
- Department of Clinical and Experimental Medicine, University of Foggia, Via Pinto 1, 71122 Foggia, Italy
| | - Domenica Mangieri
- Department of Clinical and Experimental Medicine, University of Foggia, Via Pinto 1, 71122 Foggia, Italy
| |
Collapse
|
5
|
Su J, Xia H, He H, Tang H, Zhou J, Xun Y, Liu F, Su B, Su Q. Diallyl disulfide antagonizes DJ-1 mediated proliferation, epithelial-mesenchymal transition, and chemoresistance in gastric cancer cells. ENVIRONMENTAL TOXICOLOGY 2024; 39:4105-4119. [PMID: 38642008 DOI: 10.1002/tox.24300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 02/29/2024] [Accepted: 03/31/2024] [Indexed: 04/22/2024]
Abstract
Diallyl disulfide (DADS), an organic component of allicin abstracted from garlic, possesses multi-target antitumor activity. DJ-1 performs a vital function in promoting AKT aberrant activation via down-regulating phosphatase and tensin homologue (PTEN) in tumors. It is unknown the involvement of DJ-1 in epithelial-mesenchymal transition (EMT) of gastric cancer (GC) cells. The purpose of this study is to investigate whether diallyl disulfide (DADS) intervenes in the role of DJ-1 in GC. Based on the identification that the correlation between high DJ-1 and low PTEN expression in GC was implicated in clinical progression, we illuminated that down-regulation of DJ-1 by DADS aided in an increase in PTEN expression and a decrease in phosphorylated AKT levels, which was in line with the results manifested in the DJ-1 knockdown and overexpressed cells, concurrently inhibiting proliferation, EMT, migration, and invasion. Furthermore, the antagonistic effects of DADS on DJ-1 were observed in in vivo experiments. Additionally, DADS mitigated the DJ-1-associated drug resistance. The current study revealed that DJ-1 is one of potential targets for DADS, which hopefully provides a promising strategy for prevention and adjuvant chemotherapy of GC.
Collapse
Affiliation(s)
- Jian Su
- Hunan Clinical Research Center for Gastric Cancer Prevention and Treatment, Second Affiliated hospital, University of South China, Hengyang, China
- Hunan Province Key Laboratory of Cancer Cellular and Molecular Pathology, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, China
| | - Hong Xia
- Hunan Clinical Research Center for Gastric Cancer Prevention and Treatment, Second Affiliated hospital, University of South China, Hengyang, China
- Hunan Province Key Laboratory of Cancer Cellular and Molecular Pathology, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, China
| | - Hui He
- Hunan Clinical Research Center for Gastric Cancer Prevention and Treatment, Second Affiliated hospital, University of South China, Hengyang, China
- Clinical Anatomy & Reproductive Medicine Application Institute, Hengyang Medical School, University of South China, Hengyang, China
| | - Huan Tang
- Hunan Clinical Research Center for Gastric Cancer Prevention and Treatment, Second Affiliated hospital, University of South China, Hengyang, China
- Department of Oncology, Yongzhou Central Hospital, Yongzhou, China
| | - Juan Zhou
- Hunan Clinical Research Center for Gastric Cancer Prevention and Treatment, Second Affiliated hospital, University of South China, Hengyang, China
- Hunan Province Key Laboratory of Cancer Cellular and Molecular Pathology, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, China
| | - Yi Xun
- Hunan Clinical Research Center for Gastric Cancer Prevention and Treatment, Second Affiliated hospital, University of South China, Hengyang, China
- Center for Gastric Cancer Research of Hunan Province, First Affiliated Hospital, University of South China, Hengyang, China
| | - Fang Liu
- Hunan Clinical Research Center for Gastric Cancer Prevention and Treatment, Second Affiliated hospital, University of South China, Hengyang, China
- Hunan Province Key Laboratory of Cancer Cellular and Molecular Pathology, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, China
| | - Bo Su
- Hunan Clinical Research Center for Gastric Cancer Prevention and Treatment, Second Affiliated hospital, University of South China, Hengyang, China
- Institute of Pharmacy and Pharmacology, School of Pharmacy, Hengyang Medical School, University of South China, Hengyang, China
| | - Qi Su
- Hunan Clinical Research Center for Gastric Cancer Prevention and Treatment, Second Affiliated hospital, University of South China, Hengyang, China
- Hunan Province Key Laboratory of Cancer Cellular and Molecular Pathology, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, China
| |
Collapse
|
6
|
Yavuz A, Simsek K, Alpsoy A, Altunay B, Gedik EO, Unal B, Bassorgun CI, Tatli AM, Elpek GO. Prognostic significance of tumor budding, desmoplastic reaction, and lymphocytic infiltration in patients with gastric adenocarcinoma. World J Gastrointest Pathophysiol 2024; 15:91237. [PMID: 38682027 PMCID: PMC11045359 DOI: 10.4291/wjgp.v15.i1.91237] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/25/2023] [Revised: 03/08/2024] [Accepted: 03/22/2024] [Indexed: 04/19/2024] Open
Abstract
BACKGROUND Recent studies have shown that the tumor microenvironment significantly influences the behavior of solid tumors. In this context, Accumulated data suggests that pathological evaluation of tumor budding (TB), desmoplastic reaction (DR), and tumor-infiltrating lymphocytes (TILs) may be crucial in determining tumor behavior in the gastrointestinal tract. Regarding gastric adenocarcinoma (GAC), although some results suggest that TB and TILs may be effective in determining the course of the disease, the data do not agree. Moreover, very few studies have investigated the relationship between DR and survival. At present, the associations between tumor TB, DR and TILs in GAC patients have not been determined. AIM To establish the relationships between TB, DR, and TILs in patients with GAC and to assess their influence on prognosis. METHODS Our study group comprised 130 patients diagnosed with GAC. The definition of TB was established based on the International TB Consensus Conference. The DR was categorized into three groups according to the level of tumor stroma maturation. The assessment of TILs was conducted using a semiquantitative approach, employing a cutoff value of 5%. The statistical analysis of the whole group and 100 patients with an intestinal subtype of GAC was performed using SPSS version 27. RESULTS A significant correlation between peritumoral budding (PTB) and intratumoral budding (ITB) was noted (r = 0.943). Tumors with high PTBs and ITBs had a greater incidence of immature DRs and low TILs (P < 0.01). PTB and ITB were associated with histological subtype, lymph node metastasis (LNM), and stage (P < 0.01). ITB, PTB, LNM, DR, and stage were significant risk factors associated with poor prognosis. The multivariate Cox regression analysis identified ITB, PTB, and LNM as independent prognostic variables (P < 0.05). In intestinal-type adenocarcinomas, a positive correlation between PTB and ITB was noted (r = 0.972). While univariate analysis revealed that LNM, stage, PTB, ITB, and DR were strong parameters for predicting survival (P < 0.05), only PTB and ITB were found to be independent prognostic factors (P < 0.001). CONCLUSION TB may be a potential prognostic marker in GAC. However, further studies are needed to delineate its role in pathology reporting protocols and the predictive effects of DR and TILs.
Collapse
Affiliation(s)
- Aysen Yavuz
- Department of Pathology, Akdeniz University Medical School, Antalya 07070, Türkiye
| | - Kubra Simsek
- Department of Pathology, Akdeniz University Medical School, Antalya 07070, Türkiye
| | - Anil Alpsoy
- Department of Pathology, Akdeniz University Medical School, Antalya 07070, Türkiye
| | - Busra Altunay
- Department of Pathology, Akdeniz University Medical School, Antalya 07070, Türkiye
| | - Elif Ocak Gedik
- Department of Pathology, Akdeniz University Medical School, Antalya 07070, Türkiye
| | - Betul Unal
- Department of Pathology, Akdeniz University Medical School, Antalya 07070, Türkiye
| | | | - Ali Murat Tatli
- Department of Medical Oncology, Akdeniz University Medical School, Antalya 07070, Türkiye
| | - Gulsum Ozlem Elpek
- Department of Pathology, Akdeniz University Medical School, Antalya 07070, Türkiye
| |
Collapse
|
7
|
Strippoli R, Niayesh-Mehr R, Adelipour M, Khosravi A, Cordani M, Zarrabi A, Allameh A. Contribution of Autophagy to Epithelial Mesenchymal Transition Induction during Cancer Progression. Cancers (Basel) 2024; 16:807. [PMID: 38398197 PMCID: PMC10886827 DOI: 10.3390/cancers16040807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 02/13/2024] [Accepted: 02/13/2024] [Indexed: 02/25/2024] Open
Abstract
Epithelial Mesenchymal Transition (EMT) is a dedifferentiation process implicated in many physio-pathological conditions including tumor transformation. EMT is regulated by several extracellular mediators and under certain conditions it can be reversible. Autophagy is a conserved catabolic process in which intracellular components such as protein/DNA aggregates and abnormal organelles are degraded in specific lysosomes. In cancer, autophagy plays a controversial role, acting in different conditions as both a tumor suppressor and a tumor-promoting mechanism. Experimental evidence shows that deep interrelations exist between EMT and autophagy-related pathways. Although this interplay has already been analyzed in previous studies, understanding mechanisms and the translational implications of autophagy/EMT need further study. The role of autophagy in EMT is not limited to morphological changes, but activation of autophagy could be important to DNA repair/damage system, cell adhesion molecules, and cell proliferation and differentiation processes. Based on this, both autophagy and EMT and related pathways are now considered as targets for cancer therapy. In this review article, the contribution of autophagy to EMT and progression of cancer is discussed. This article also describes the multiple connections between EMT and autophagy and their implication in cancer treatment.
Collapse
Affiliation(s)
- Raffaele Strippoli
- Department of Molecular Medicine, Sapienza University of Rome, 00161 Rome, Italy;
- National Institute for Infectious Diseases “Lazzaro Spallanzani”, I.R.C.C.S., 00149 Rome, Italy
| | - Reyhaneh Niayesh-Mehr
- Department of Clinical Biochemistry, Faculty of Medical Science, Tarbiat Modares University, Tehran P.O. Box 14115-331, Iran;
| | - Maryam Adelipour
- Department of Clinical Biochemistry, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz 61357-15794, Iran;
| | - Arezoo Khosravi
- Department of Genetics and Bioengineering, Faculty of Engineering and Natural Sciences, Istanbul Okan University, Istanbul 34959, Türkiye;
| | - Marco Cordani
- Department of Biochemistry and Molecular Biology, Faculty of Biological Sciences, Complutense University of Madrid, 28040 Madrid, Spain;
- Instituto de Investigaciones Sanitarias San Carlos (IdISSC), 28040 Madrid, Spain
| | - Ali Zarrabi
- Department of Biomedical Engineering, Faculty of Engineering and Natural Sciences, Istinye University, Istanbul 34396, Türkiye;
- Department of Research Analytics, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai 600077, India
| | - Abdolamir Allameh
- Department of Clinical Biochemistry, Faculty of Medical Science, Tarbiat Modares University, Tehran P.O. Box 14115-331, Iran;
| |
Collapse
|
8
|
Odarenko KV, Zenkova MA, Markov AV. The Nexus of Inflammation-Induced Epithelial-Mesenchymal Transition and Lung Cancer Progression: A Roadmap to Pentacyclic Triterpenoid-Based Therapies. Int J Mol Sci 2023; 24:17325. [PMID: 38139154 PMCID: PMC10743660 DOI: 10.3390/ijms242417325] [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: 11/01/2023] [Revised: 12/06/2023] [Accepted: 12/07/2023] [Indexed: 12/24/2023] Open
Abstract
Lung cancer is the leading cause of cancer-related death worldwide. Its high mortality is partly due to chronic inflammation that accompanies the disease and stimulates cancer progression. In this review, we analyzed recent studies and highlighted the role of the epithelial-mesenchymal transition (EMT) as a link between inflammation and lung cancer. In the inflammatory tumor microenvironment (iTME), fibroblasts, macrophages, granulocytes, and lymphocytes produce inflammatory mediators, some of which can induce EMT. This leads to increased invasiveness of tumor cells and self-renewal of cancer stem cells (CSCs), which are associated with metastasis and tumor recurrence, respectively. Based on published data, we propose that inflammation-induced EMT may be a potential therapeutic target for the treatment of lung cancer. This prospect is partially realized in the development of EMT inhibitors based on pentacyclic triterpenoids (PTs), described in the second part of our study. PTs reduce the metastatic potential and stemness of tumor cells, making PTs promising candidates for lung cancer therapy. We emphasize that the high diversity of molecular mechanisms underlying inflammation-induced EMT far exceeds those that have been implicated in drug development. Therefore, analysis of information on the relationship between the iTME and EMT is of great interest and may provide ideas for novel treatment approaches for lung cancer.
Collapse
Affiliation(s)
- Kirill V. Odarenko
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, 630090 Novosibirsk, Russia; (K.V.O.); (M.A.Z.)
- Faculty of Natural Sciences, Novosibirsk State University, 630090 Novosibirsk, Russia
| | - Marina A. Zenkova
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, 630090 Novosibirsk, Russia; (K.V.O.); (M.A.Z.)
| | - Andrey V. Markov
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, 630090 Novosibirsk, Russia; (K.V.O.); (M.A.Z.)
| |
Collapse
|
9
|
Chen Y, Wang X, Ye D, Yang Z, Shen Q, Liu X, Chen C, Chen X. Research progress of sophoridine's pharmacological activities and its molecular mechanism: an updated review. Front Pharmacol 2023; 14:1126636. [PMID: 37397472 PMCID: PMC10311568 DOI: 10.3389/fphar.2023.1126636] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Accepted: 06/07/2023] [Indexed: 07/04/2023] Open
Abstract
Background: Sophoridine, the major active constituent of Sophora alopecuroides and its roots, is a bioactive alkaloid with a wide range of pharmacological effects, including antitumor, anti-inflammatory, antiviral, antibacterial, analgesic, cardioprotective, and immunoprotective activities. Sophora flavescens Aiton is a traditional Chinese medicine that is bitter and cold. Additionally, it also exhibits the effects of clearing heat, eliminating dampness, and expelling insects. Aims of the study: To summarize the pharmacological research and associated mechanisms of sophoridine, we compiled this review by combining a huge body of relevant literature. Materials and methods: The information related to this article was systematically collected from the scientific literature databases including PubMed, Google Scholar, Web of Science, Science Direct, Springer, China National Knowledge Infrastructure, published books, PhD and MS dissertations. Results: Its antitumor activity is particularly remarkable, as it can inhibit cancer cell proliferation, invasion, and metastasis while inducing cell cycle arrest and apoptosis. Additionally, sophoridine also holds therapeutic potential for myocardial ischemia, osteoporosis, arrhythmias, and neurological disorders, primarily through the suppression of related inflammatory factors and cell apoptosis. However, sophoridine has also exhibited adverse effects such as hepatotoxicity and neurotoxicity. The antidisease effect and mechanism of sophoridine are diverse, so it has high research value. Conclusion: As an important traditional Chinese medicine alkaloid, modern pharmacological studies have demonstrated that sophoridine has prominent bioactivities, especially on anti-tumor anti-inflammation activities, and cardiovascular system protection. These activities provide prospects for novel drug development for cancer and some chronic diseases. Nevertheless, the understanding of the multitarget network pharmacology, long-term in vivo toxicity, and clinical efficacy of sophoridine require further detailed research.
Collapse
Affiliation(s)
- Yiwei Chen
- Department of Pharmacy, Guangxi Academy of Medical Sciences and the People’s Hospital of Guangxi Zhuang Autonomous, Nanning, China
| | - Xiang Wang
- School of Chinese Meteria Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Dongmei Ye
- Department of Pharmacy, Guangxi Academy of Medical Sciences and the People’s Hospital of Guangxi Zhuang Autonomous, Nanning, China
| | - Zhousheng Yang
- Department of Pharmacy, Guangxi Academy of Medical Sciences and the People’s Hospital of Guangxi Zhuang Autonomous, Nanning, China
| | - Qingrong Shen
- Department of Pharmacy, Guangxi Academy of Medical Sciences and the People’s Hospital of Guangxi Zhuang Autonomous, Nanning, China
| | - Xiaoxia Liu
- Department of Pharmacy, Guangxi Academy of Medical Sciences and the People’s Hospital of Guangxi Zhuang Autonomous, Nanning, China
| | - Chunxia Chen
- Department of Pharmacy, Guangxi Academy of Medical Sciences and the People’s Hospital of Guangxi Zhuang Autonomous, Nanning, China
| | - Xiaoyu Chen
- Department of Pharmacy, Guangxi Academy of Medical Sciences and the People’s Hospital of Guangxi Zhuang Autonomous, Nanning, China
| |
Collapse
|
10
|
Zhang C, Zhang Y, Hu X, Zhao Z, Chen Z, Wang X, Zhang Z, Jin H, Zhang J. Luteolin inhibits subretinal fibrosis and epithelial-mesenchymal transition in laser-induced mouse model via suppression of Smad2/3 and YAP signaling. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 116:154865. [PMID: 37201365 DOI: 10.1016/j.phymed.2023.154865] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 04/13/2023] [Accepted: 05/08/2023] [Indexed: 05/20/2023]
Abstract
BACKGROUND Subretinal fibrosis (SF) accounts for vision loss in patients with neovascular age-related macular degeneration (nAMD) even treated with adequate intravitreal injection of anti-vascular endothelial growth factor (anti-VEGF) drugs. Currently, there is no treatment available to prevent or treat SF caused by nAMD. PURPOSE This study aims to investigate the potential effects of luteolin on SF and epithelial-mesenchymal transition (EMT) as well as the underlying molecular pathways both in vivo and in vitro. METHODS Seven-week-old male C57BL/6J mice were employed to establish laser-induced choroidal neovascularization (CNV) and SF. One day after the laser induction, luteolin was administered intravitreally. SF and CNV were assessed with the immunolabeling of collagen type I (collagen I) and isolectin B4 (IB4), respectively. RPE65 and α-SMA colocalization in the lesions was used to evaluate the extent of EMT in retinal pigment epithelial (RPE) cells by using immunofluorescence. In vitro, luteolin was administered to TGFβ1-treated primary human RPE (phRPE) cells. RT-qPCR, Western blot and immunofluorescence were employed to evaluate the change of EMT-related molecules, epithelial markers, and relevant signaling pathways. The functional changes associated with EMT were investigated using the scratch assay, Transwell migration assay, and collagen gel contraction assay. CCK-8 was used to determine the cell viability of phRPE cells. RESULTS On day 7 and 14 after laser induction in mice, intravitreal injection of luteolin dramatically decreased the immunolabeled sizes of both collagen I and IB4, as well as the amount of colocalized double immunostaining of α-SMA and RPE65 in laser-induced SF lesions. In vitro, TGFβ1-treated phRPE cells demonstrated increased cell migration and contraction capacity, accompanied with considerable overexpression of fibronectin, α-SMA, N-cadherin and vimentin, as well as downregulation of E-cadherin and ZO-1. The above changes were largely inhibited by luteolin co-incubation. Mechanistically, luteolin could evidently decrease the phosphorylation of Smad2/3, whereas increase the phosphorylation of YAP in TGFβ1-treated phRPE cells. CONCLUSION This study demonstrates that luteolin exhibits the anti-fibrotic effect in a laser-induced mouse model by inhibiting EMT of RPE cells via deactivating Smad2/3 and YAP signaling, which provides a potential natural compound for the prevention and treatment of SF and fibrosis-related diseases.
Collapse
Affiliation(s)
- Chaoyang Zhang
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People's Hospital), Shanghai Jiao Tong University School of Medicine, Shanghai, PR China; National Clinical Research Center for Eye Diseases, Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, PR China
| | - Yao Zhang
- Department of Ophthalmology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, PR China
| | - Xin Hu
- Department of Ophthalmology, Huaihe Hospital, Henan University, Kaifeng, PR China
| | - Zhenzhen Zhao
- Department of Regenerative Medicine, and Department of Pharmacology, Tongji University School of Medicine, Shanghai, PR China
| | - Ziang Chen
- Department of Ophthalmology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, PR China
| | - Xi Wang
- Department of Regenerative Medicine, and Department of Pharmacology, Tongji University School of Medicine, Shanghai, PR China
| | - Zhihua Zhang
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People's Hospital), Shanghai Jiao Tong University School of Medicine, Shanghai, PR China; National Clinical Research Center for Eye Diseases, Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, PR China
| | - Haiying Jin
- Department of Ophthalmology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, PR China.
| | - Jingfa Zhang
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People's Hospital), Shanghai Jiao Tong University School of Medicine, Shanghai, PR China; National Clinical Research Center for Eye Diseases, Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, PR China.
| |
Collapse
|
11
|
Gahtori R, Tripathi AH, Kumari A, Negi N, Paliwal A, Tripathi P, Joshi P, Rai RC, Upadhyay SK. Anticancer plant-derivatives: deciphering their oncopreventive and therapeutic potential in molecular terms. FUTURE JOURNAL OF PHARMACEUTICAL SCIENCES 2023. [DOI: 10.1186/s43094-023-00465-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/25/2023] Open
Abstract
Abstract
Background
Over the years, phytomedicines have been widely used as natural modalities for the treatment and prevention of various diseases by different ethnic groups across the globe. Although, 25% of drugs in the USA contain at least one plant-derived therapeutic compound, currently there is a paucity of plant-derived active medicinal ingredients in the pharmaceutical industry. Scientific evidence-based translation of plant-derived ethnomedicines for their clinical application is an urgent need. The anticancer and associated properties (antioxidative, anti-inflammatory, pro-apoptotic and epithelial-mesenchymal transition (EMT) inhibition) of various plant extracts and phytochemicals have been elucidated earlier. Several of the plant derivatives are already in use under prophylactic/therapeutic settings against cancer and many are being investigated under different phases of clinical trials.
Main body
The purpose of this study is to systematically comprehend the progress made in the area of prophylactic and therapeutic potential of the anticancerous plant derivatives. Besides, we aim to understand their anticancer potential in terms of specific sub-phenomena, such as anti-oxidative, anti-inflammatory, pro-apoptotic and inhibition of EMT, with an insight of the molecules/pathways associated with them. The study also provides details of classes of anticancer compounds, their plant source(s) and the molecular pathway(s) targeted by them. In addition to the antioxidative and antiproliferative potentials of anticancer plant derivatives, this study emphasizes on their EMT-inhibition potential and other ‘anticancer related’ properties. The EMT is highlighted as a phenomenon of choice for targeting cancer due to its role in the induction of metastasis and drug resistance. Different phytochemicals in pre-clinical or clinical trials, with promising chemopreventive/anticancer activities have been enlisted and the plant compounds showing synergistic anticancer activity in combination with the existing drugs have been discussed. The review also unravels the need of carrying out pan-signalome studies for identifying the cardinal pathways modulated by phytomedicine(s), as in many cases, the molecular pathway(s) has/have been randomly studied.
Conclusion
This review systematically compiles the studies regarding the impact of various plant derivatives in different cancers and oncogenic processes, as tested in diverse experimental model systems. Availability of more comprehensive information on anticancer phyto-constituents, their relative abundance in crude drugs, pathways/molecules targeted by phytomedicines, their long-term toxicity data and information regarding their safe use under the combinatorial settings, would open greater avenues of their utilization in future against this dreaded disease.
Graphical Abstract
Collapse
|
12
|
Wu F, Liu Y, Hu S, Lu C. Ribosomal protein L31 (RPL31) inhibits the proliferation and migration of gastric cancer cells. Heliyon 2023; 9:e13076. [PMID: 36816257 PMCID: PMC9936522 DOI: 10.1016/j.heliyon.2023.e13076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 01/04/2023] [Accepted: 01/16/2023] [Indexed: 01/21/2023] Open
Abstract
Gastric cancer (GC) is a digestive tract malignant tumor and causes the third cancer-related mortality in the world. Aberrant expression of Ribosomal Protein L31 (RPL31) has been reported in several human cancers. The aim of this study was to explore the role and possible biological functions of RPL31 in GC. We firstly employed immunohistochemistry to examine RPL31 expression in tumor and para-cancerous tissues. By lentiviral transfection, we successfully constructed an RPL31-knockdown GC cell model and performed functional validation to reveal the effects of RPL31 on proliferation, apoptosis, cycle, migration, and tumor growth. Our data indicated that RPL31 was abundantly expressed in GC tissues and cell lines (AGS and MGC-803). In addition, RPL31 expression was positively correlated with the extent of tumor infiltrate of GC patients. Functionally, silencing RPL31 in AGS and MGC-803 cells significantly limited the ability of proliferation and migration, promoted cell apoptosis. Consistently, RPL31-knockdown GC cells inhibited the growth of xenografts in mice. Moreover, preliminary analysis on the downstream regulation mechanism revealed that RPL31 functioned as a tumor promoter through targeting JAK-STAT signaling pathway. In conclusion, inhibition of abnormally high expression of RPL31 in GC may be a potential therapeutic strategy for this disease.
Collapse
Affiliation(s)
- Fang Wu
- Department of Oncology, The First Affiliated Hospital of Nanchang University. Yongwaizheng Street, No. 17, Nanchang City, Jiangxi Province 330006, China
| | - Yangyang Liu
- Department of Oncology, The First Affiliated Hospital of Nanchang University. Yongwaizheng Street, No. 17, Nanchang City, Jiangxi Province 330006, China
| | - Shenglin Hu
- Department of Internal Medicine, People’s Hospital of Jinan County. Beida Street, No. 5, Jinan City, Jiangxi Province 330699, China
| | - Canrong Lu
- Senior Department of General Surgery, The First Medical Center of Chinese, PLA General Hospital. Fuxin Road, No. 28, Haidian District, Beijing 100853, China,Corresponding author.
| |
Collapse
|
13
|
Duan X, Luo M, Li J, Shen Z, Xie K. Overcoming therapeutic resistance to platinum-based drugs by targeting Epithelial–Mesenchymal transition. Front Oncol 2022; 12:1008027. [PMID: 36313710 PMCID: PMC9614084 DOI: 10.3389/fonc.2022.1008027] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Accepted: 09/23/2022] [Indexed: 11/30/2022] Open
Abstract
Platinum-based drugs (PBDs), including cisplatin, carboplatin, and oxaliplatin, have been widely used in clinical practice as mainstay treatments for various types of cancer. Although there is firm evidence of notable achievements with PBDs in the management of cancers, the acquisition of resistance to these agents is still a major challenge to efforts at cure. The introduction of the epithelial-mesenchymal transition (EMT) concept, a critical process during embryonic morphogenesis and carcinoma progression, has offered a mechanistic explanation for the phenotypic switch of cancer cells upon PBD exposure. Accumulating evidence has suggested that carcinoma cells can enter a resistant state via induction of the EMT. In this review, we discussed the underlying mechanism of PBD-induced EMT and the current understanding of its role in cancer drug resistance, with emphasis on how this novel knowledge can be exploited to overcome PBD resistance via EMT-targeted compounds, especially those under clinical trials.
Collapse
Affiliation(s)
- Xirui Duan
- Department of Oncology, Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Maochao Luo
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Jian Li
- West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China
| | - Zhisen Shen
- Department of Otorhinolaryngology and Head and Neck Surgery, The Affiliated Lihuili Hospital, Ningbo University, Ningbo, China
- *Correspondence: Ke Xie, ; Zhisen Shen,
| | - Ke Xie
- Department of Oncology, Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
- *Correspondence: Ke Xie, ; Zhisen Shen,
| |
Collapse
|
14
|
Wang B, Zou D, Wang N, Wang H, Zhang T, Gao L, Ma C, Zheng P, Gu B, Li X, Wang Y, He P, Ma Y, Wang X, Chen H. Construction and validation of a novel coagulation-related 7-gene prognostic signature for gastric cancer. Front Genet 2022; 13:957655. [PMID: 36105100 PMCID: PMC9465170 DOI: 10.3389/fgene.2022.957655] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 07/25/2022] [Indexed: 11/18/2022] Open
Abstract
Background: Gastric cancer (GC) is the most common malignant tumor. Due to the lack of practical molecular markers, the prognosis of patients with advanced gastric cancer is still poor. A number of studies have confirmed that the coagulation system is closely related to tumor progression. Therefore, the purpose of this study was to construct a coagulation-related gene signature and prognostic model for GC by bioinformatics methods. Methods: We downloaded the gene expression and clinical data of GC patients from the TCGA and GEO databases. In total, 216 coagulation-related genes (CRGs) were obtained from AmiGO 2. Weighted gene co-expression network analysis (WGCNA) was used to identify coagulation-related genes associated with the clinical features of GC. Last absolute shrinkage and selection operator (LASSO) Cox regression was utilized to shrink the relevant predictors of the coagulation system, and a Coag-Score prognostic model was constructed based on the coefficients. According to this risk model, GC patients were divided into high-risk and low-risk groups, and overall survival (OS) curves and receiver operating characteristic (ROC) curves were drawn in the training and validation sets, respectively. We also constructed nomograms for predicting 1-, 2-, and 3-year survival in GC patients. Single-sample gene set enrichment analysis (ssGSEA) was exploited to explore immune cells’ underlying mechanisms and correlations. The expression levels of coagulation-related genes were verified by real-time quantitative polymerase chain reaction (qRT-PCR) and immunohistochemistry (IHC). Results: We identified seven CRGs employed to construct a Coag-Score risk model using WGCNA combined with LASSO regression. In both training and validation sets, GC patients in the high-risk group had worse OS than those in the low-risk group, and Coag-Score was identified as an independent predictor of OS, and the nomogram provided a quantitative method to predict the 1-, 2-, and 3-year survival rates of GC patients. Functional analysis showed that Coag-Score was mainly related to the MAPK signaling pathway, complement and coagulation cascades, angiogenesis, epithelial–mesenchymal transition (EMT), and KRAS signaling pathway. In addition, the high-risk group had a significantly higher infiltration enrichment score and was positively associated with immune checkpoint gene expression. Conclusion: Coagulation-related gene models provide new insights and targets for the diagnosis, prognosis prediction, and treatment management of GC patients.
Collapse
Affiliation(s)
- Bofang Wang
- Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Dan Zou
- Chengdu Seventh People’s Hospital, Chengdu, China
| | - Na Wang
- Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Haotian Wang
- State Key Laboratory of Genetic Resources and Evolution/Key Laboratory of Healthy Aging Research of Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Tao Zhang
- Second Clinical Medical College, Lanzhou University, Lanzhou, China
- Department of oncology, First Hospital of Lanzhou University, Lanzhou, China
| | - Lei Gao
- Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Chenhui Ma
- Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Peng Zheng
- Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Baohong Gu
- Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Xuemei Li
- Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Yunpeng Wang
- Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Puyi He
- Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Yanling Ma
- Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Xueyan Wang
- Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Hao Chen
- Second Clinical Medical College, Lanzhou University, Lanzhou, China
- Key Laboratory of the Digestive System Tumors of Gansu Province, Lanzhou, China
- Department of Cancer Center, Lanzhou University Second Hospital, Lanzhou, China
- *Correspondence: Hao Chen,
| |
Collapse
|
15
|
Xiaotan Sanjie Decoction Inhibits Gastric Cancer Cell Proliferation, Migration, and Invasion through lncRNA-ATB and miR-200A. BIOMED RESEARCH INTERNATIONAL 2022; 2022:7029182. [PMID: 36060143 PMCID: PMC9436559 DOI: 10.1155/2022/7029182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 07/08/2022] [Accepted: 07/11/2022] [Indexed: 11/24/2022]
Abstract
This study is aimed at exploring whether Xiaotan Sanjie decoction (XTSJ) inhibits gastric cancer (GC) proliferation and metastasis by regulating lncRNA-ATB expression. qRT-PCR and Western blot were used to analyze lncRNA-ATB and downstream-regulated genes/proteins in human GC cells. CCK8, Edu, and flow cytometry assays were used to detect the inhibitory effect of XTSJ on cell proliferation and apoptosis. Moreover, transwell and wound healing assays were used to detect the inhibitory effect of XTSJ on migration and invasion. qRT-PCR and Western blot were used to detect regulated genes and proteins levels. The HGC-27 cell line was used for follow-up analysis due to the high level of lncRNA-ATB and cell characteristics. XTSJ inhibited the proliferation and metastasis of HGC-27 in a dose-dependent manner. Further research found that XTSJ downregulated lncRNA-ATB, Vimentin, and N-cadherin, while it upregulated miR-200a and E-cadherin in a dose-dependent manner. XTSJ also upregulated Caspase 3, Caspase 9, Bax, and downregulated Bcl-2. Furthermore, XTSJ inhibited tumor growth in vivo and downregulated EMT signaling pathways. These results indicate that XTSJ may affect EMT and Bcl-2 signaling pathways by regulating lncRNA-ATB and miR-200a, thus inhibiting proliferation, migration, and invasion of HGC-27 cells. Therefore, XTSJ may be an effective treatment for the high levels of lncRNA-ATB in GC.
Collapse
|
16
|
Li X, Chen L, Peng X, Zhan X. Progress of tumor-associated macrophages in the epithelial-mesenchymal transition of tumor. Front Oncol 2022; 12:911410. [PMID: 35965509 PMCID: PMC9366252 DOI: 10.3389/fonc.2022.911410] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Accepted: 07/07/2022] [Indexed: 11/29/2022] Open
Abstract
As a significant public health problem with high morbidity and mortality worldwide, tumor is one of the major diseases endangering human life. Moreover, metastasis is the most important contributor to the death of tumor patients. Epithelial-mesenchymal transition (EMT) is an essential biological process in developing primary tumors to metastasis. It underlies tumor progression and metastasis by inducing a series of alterations in tumor cells that confer the ability to move and migrate. Tumor-associated macrophages (TAMs) are one of the primary infiltrating immune cells in the tumor microenvironment, and they play an indispensable role in the EMT process of tumor cells by interacting with tumor cells. With the increasing clarity of the relationship between TAMs and EMT and tumor metastasis, targeting TAMs and EMT processes is emerging as a promising target for developing new cancer therapies. Therefore, this paper reviews the recent research progress of tumor-associated macrophages in tumor epithelial-mesenchymal transition and briefly discusses the current anti-tumor therapies targeting TAMs and EMT processes.
Collapse
Affiliation(s)
| | | | - Xiaobo Peng
- *Correspondence: Xiaobo Peng, ; Xianbao Zhan,
| | | |
Collapse
|
17
|
Resveratrol inhibits TGF-β1-induced EMT in gastric cancer cells through Hippo-YAP signaling pathway. Clin Transl Oncol 2022; 24:2210-2221. [DOI: 10.1007/s12094-022-02882-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 06/24/2022] [Indexed: 11/25/2022]
|
18
|
Tanabe S, O’Brien J, Tollefsen KE, Kim Y, Chauhan V, Yauk C, Huliganga E, Rudel RA, Kay JE, Helm JS, Beaton D, Filipovska J, Sovadinova I, Garcia-Reyero N, Mally A, Poulsen SS, Delrue N, Fritsche E, Luettich K, La Rocca C, Yepiskoposyan H, Klose J, Danielsen PH, Esterhuizen M, Jacobsen NR, Vogel U, Gant TW, Choi I, FitzGerald R. Reactive Oxygen Species in the Adverse Outcome Pathway Framework: Toward Creation of Harmonized Consensus Key Events. FRONTIERS IN TOXICOLOGY 2022; 4:887135. [PMID: 35875696 PMCID: PMC9298159 DOI: 10.3389/ftox.2022.887135] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Accepted: 06/13/2022] [Indexed: 02/05/2023] Open
Abstract
Reactive oxygen species (ROS) and reactive nitrogen species (RNS) are formed as a result of natural cellular processes, intracellular signaling, or as adverse responses associated with diseases or exposure to oxidizing chemical and non-chemical stressors. The action of ROS and RNS, collectively referred to as reactive oxygen and nitrogen species (RONS), has recently become highly relevant in a number of adverse outcome pathways (AOPs) that capture, organize, evaluate and portray causal relationships pertinent to adversity or disease progression. RONS can potentially act as a key event (KE) in the cascade of responses leading to an adverse outcome (AO) within such AOPs, but are also known to modulate responses of events along the AOP continuum without being an AOP event itself. A substantial discussion has therefore been undertaken in a series of workshops named "Mystery or ROS" to elucidate the role of RONS in disease and adverse effects associated with exposure to stressors such as nanoparticles, chemical, and ionizing and non-ionizing radiation. This review introduces the background for RONS production, reflects on the direct and indirect effects of RONS, addresses the diversity of terminology used in different fields of research, and provides guidance for developing a harmonized approach for defining a common event terminology within the AOP developer community.
Collapse
Affiliation(s)
- Shihori Tanabe
- Division of Risk Assessment, Center for Biological Safety and Research, National Institute of Health Sciences, Kawasaki, Japan
| | - Jason O’Brien
- Wildlife Toxicology Research Section, Environment and Climate Change Canada, Toronto, ON, Canada
| | - Knut Erik Tollefsen
- Norwegian Institute for Water Research (NIVA), Oslo, Norway
- Norwegian University of Life Sciences (NMBU), Ås, Norway
- Centre for Environmental Radioactivity, Norwegian University of Life Sciences (NMBU), Ås, Norway
| | - Youngjun Kim
- Korea Institute of Science and Technology (KIST) Europe, Saarbrücken, Germany
| | | | | | | | | | | | | | | | | | - Iva Sovadinova
- RECETOX, Faculty of Science, Masaryk University, Brno, Czech Republic
| | - Natalia Garcia-Reyero
- U.S. Army Engineer Research and Development Center (ERDC), Vicksburg, MS, United States
| | - Angela Mally
- Department of Toxicology, University of Würzburg, Würzburg, Germany
| | - Sarah Søs Poulsen
- National Research Centre for the Working Environment, Copenhagen, Denmark
| | - Nathalie Delrue
- Organisation for Economic Co-operation and Development (OECD), Paris, France
| | - Ellen Fritsche
- Group of Alternative Method Development for Environmental Toxicity Testing, IUF—Leibniz-Research Institute for Environmental Medicine, Duesseldorf, Germany
| | - Karsta Luettich
- Philip Morris International R&D, Philip Morris Products SA, Neuchatel, Switzerland
| | - Cinzia La Rocca
- Center for Gender-specific Medicine, Italian National Institute of Health, Rome, Italy
| | - Hasmik Yepiskoposyan
- Philip Morris International R&D, Philip Morris Products SA, Neuchatel, Switzerland
| | - Jördis Klose
- Group of Alternative Method Development for Environmental Toxicity Testing, IUF—Leibniz-Research Institute for Environmental Medicine, Duesseldorf, Germany
| | | | - Maranda Esterhuizen
- University of Helsinki, Ecosystems and Environment Research Programme, Faculty of Biological and Environmental Sciences, Lahti, Finland, and Helsinki Institute of Sustainability Science (HELSUS), Helsinki, Finland
| | | | - Ulla Vogel
- National Research Centre for the Working Environment, Copenhagen, Denmark
| | - Timothy W. Gant
- UK Health Security Agency, Public Health England, London, United Kingdom
| | - Ian Choi
- Korea Institute of Science and Technology (KIST) Europe, Saarbrücken, Germany
| | | |
Collapse
|
19
|
Upregulated Circular RNA KIF4A Promotes Cell Migration and Invasion by Regulating MicroRNA-144-3p/EZH2 Axis in Gastric Cancer. JOURNAL OF ONCOLOGY 2022; 2022:3985621. [PMID: 35466313 PMCID: PMC9033408 DOI: 10.1155/2022/3985621] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 03/16/2022] [Indexed: 12/15/2022]
Abstract
Accumulating evidence has shown that circular RNAs (circRNAs) serve a critical regulatory role in various human cancers, including gastric cancer (GC), and in this study, we aimed to explore the functions of circKIF4A in the progression of GC. Our findings demonstrated that circKIF4A was highly expressed in both GC tissues and cell lines, and high intratumoral circKIF4A expression predicted a poor prognosis in GC patients. In vitro gain- and loss-of-function assays indicated that circKIF4A knockdown suppressed the proliferation, migration, invasion, and EMT of GC cells, while these malignant behaviors were enhanced by circKIF4A overexpression. Mechanistically, we found that circKIF4A was mainly located in the cytoplasm, could directly interact with microRNA- (miR-) 144-3p, and functions as a miRNA sponge to regulate EZH2 expression in GC cells. miR-144-3p inhibition or EZH2 restoration largely blocked the effects of circKIF4A knockdown on the malignant behaviors of GC cells. This study indicated that circKIF4A can efficiently sponge miR-144-3p to promote the malignant behaviors of GC cells and may provide a potential biomarker and therapeutic target for GC management.
Collapse
|
20
|
Liu N, Jiang F, Ye M, Wang B, Ge D, Chang S. HuR confers IL-17a-induced migration and invasion of gastric cancer cells via upregulation of Snail translation. Cytokine 2022; 153:155830. [PMID: 35247650 DOI: 10.1016/j.cyto.2022.155830] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 02/07/2022] [Accepted: 02/08/2022] [Indexed: 02/07/2023]
Abstract
Human gastric cancer is a leading cause of cancer mortality in the world wide. We found that the expression of IL-17a was significantly increased in gastric cancer cells. Treatment with recombinant IL-17a (rIL-17a) can increase migration, invasion and epithelial to mesenchymal transition (EMT) of gastric cancer cells. Further, Snail, a key factor to regulate EMT, was significantly increased in rIL-17a-treated gastric cancer cells. While knockdown of Snail can abolish IL-17a-induced EMT of gastric cancer cells. Mechanistically, IL-17a can promote the translation efficiency of Snail, while had no effect on its mRNA expression or protein stability. Further, we found that IL-17a can increase the expression of HuR, which markedly promoted translation of Snail mRNA. While knockdown of HuR can reverse rIL-17a-induced expression of Snail and EMT of gastric cancer cells. Collectively, our data suggested that HuR confers IL-17a induced migration and invasion of gastric cancer cells via upregulation of Snail translation.
Collapse
Affiliation(s)
- Ning Liu
- Department of Gastrointestinal Surgery, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou 570311, Hainan Province, China
| | - Fan Jiang
- Center of Gerontology, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou 570311, Hainan Province, China
| | - Mulin Ye
- Department of Gastrointestinal Surgery, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou 570311, Hainan Province, China
| | - Bangjie Wang
- Department of Gastrointestinal Surgery, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou 570311, Hainan Province, China
| | - Dongsheng Ge
- Department of Gastrointestinal Surgery, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou 570311, Hainan Province, China
| | - Shunwu Chang
- Department of Gastrointestinal Surgery, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou 570311, Hainan Province, China.
| |
Collapse
|
21
|
Expression of tight junction transmembrane protein Claudin-1 in gastric carcinoma and effects on tumor cell proliferation, invasion and migration. REV ROMANA MED LAB 2022. [DOI: 10.2478/rrlm-2022-0007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Abstract
Background: Claudin-1 is involved in various cancers, but its expression and role in gastric carcinoma remain unclear.
Materials and Methods: Gastric carcinoma and adjacent normal tissues were harvested from 60 patients. Claudin-1 expression was detected by RT-qPCR. The expressions in human gastric carcinoma MKN45, SGC7901 and MKN28 cells and immortalized human gastric epithelium GES-1 cells were determined by RT-qPCR and Western blotting. Claudin-1 was overexpressed in SGC7901 cells by lentiviral transfection, and they were divided into Control (untransfected), normal control (NC) (transfected with lentiviral vector) and Claudin-1 (transfected with Claudin-1 overexpression lentivirus) groups. The proliferation, invasion and migration of gastric carcinoma cells were detected through cell counting kit-8, Transwell and wound healing assays, respectively. The effects of Claudin-1 on the expressions of epithelial-mesenchymal transition (EMT) marker proteins E-cadherin and N-cadherin were detected by Western blotting. Ten 4-week-old male BALB/c nude mice were subcutaneously injected with lentivirus-treated SGC7901 cells to establish the transplanted tumor model, and the effect of overexpression of Claudin-1 was explored.
Results: The expression of Claudin-1 in gastric carcinoma tissues was significantly lower than that in adjacent tissues (P<0.05). Overexpression of Claudin-1 significantly inhibited the proliferation, invasion and migration of SGC7901 cells, increased the expression of E-cadherin, and decreased that of N-cadherin (P<0.05). Overexpression of Claudin-1 in the mouse model significantly inhibited the growth of subcutaneous transplanted tumors (P<0.05).
Conclusion: Claudin-1 has low expression in gastric carcinoma tissues. Overexpression of Claudin-1 inhibits the proliferation, invasion, migration, and EMT of gastric carcinoma cells, and subcutaneous tumorigenesis in nude mice.
Collapse
|
22
|
Zhang T, Zheng J, Qiao L, Zhao W. Deubiquitinase USP13 promotes the epithelial-mesenchymal transition and metastasis in gastric cancer by maintaining Snail protein. Pathol Res Pract 2021; 229:153705. [PMID: 34872023 DOI: 10.1016/j.prp.2021.153705] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 09/13/2021] [Accepted: 11/20/2021] [Indexed: 12/12/2022]
Abstract
The dynamic balance between ubiquitination and deubiquitination is a key mechanism that regulates protein degradation and maintains cell protein homeostasis. Ubiquitin-specific peptidase 13 (USP13), a deubiquitinase (DUB), regulates various physiological and pathological processes, including cancer. A previous study reported that high USP13 mRNA expression confers poor prognosis in gastric cancer (GC). However, the biological function of USP13 in GC remains unknown. Here, we revealed that USP13 expression was upregulated in GC tissue samples compared to noncancerous tissues. USP13-positive expression was associated with poor differentiation, high invasiveness, and advanced tumor stage. Notably, upregulated USP13 expression was closely correlated with the reduced survival of GC patients. We also confirmed increased USP13 expression in GC cell lines. USP13 knockdown prominently suppressed MGC-803 cell migration and invasion. Conversely, USP13 overexpression markedly enhanced SGC-7901 cell motility. Furthermore, USP13 positively regulates the epithelial-mesenchymal transition (EMT) of GC cells. Interestingly, USP13 remarkably enhanced Snail protein expression but did not affect its mRNA levels in GC cells. We confirmed a positive correlation between USP13 and Snail expression in GC tissues. Mechanistically, USP13 knockdown promoted Snail degradation, which could be blocked by the proteasome inhibitor MG132. USP13 interacted with Snail to deubiquitinate and stabilize Snail in GC cells. Finally, Snail knockdown significantly blocked USP13-induced SGC-7901 cell migration and invasion. In conclusion, USP13 overexpression was frequently detected in GC and contributed to the EMT and metastasis of GC by stabilizing Snail.
Collapse
Affiliation(s)
- Ting Zhang
- Department of Outpatient, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Jianbao Zheng
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Lina Qiao
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China.
| | - Wei Zhao
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China.
| |
Collapse
|
23
|
周 立, 吴 忧, 辛 林. [Expression characteristics and functional analysis of ELK3 in gastric cancer]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2021; 41:1287-1295. [PMID: 34658341 PMCID: PMC8526318 DOI: 10.12122/j.issn.1673-4254.2021.09.01] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Indexed: 11/24/2022]
Abstract
OBJECTIVE To explore the expression characteristics of ELK3 and its role in the occurrence, progression and prognosis of gastric cancer. METHODS We analyzed the expression characteristics of ELK3 in gastric cancer based on E-MTAB-6693 dataset and explored the prognostic value of ELK3 using Kaplan-Meier survival analysis and univariate and multivariate Cox regression analysis. Chip-Atlas, ChipBase, Genes Transcription Regulation Database, and hTFtarget were used for predicting the target genes of ELK3 and constructing the transcription regulation network. Functional enrichment analysis of the target genes was performed using R software. The proportions of infiltrating immune cells in gastric cancer were analyzed using Cibersort tool, and the Pearson coefficients between ELK3 and these cells were calculated. The expression profile of ELK3 was verified based on Gene Expression Profiling Interactive Analysis and Human Protein Atlas databases. We also collected 5 pairs of gastric cancer and adjacent tissue samples and detected the expression of ELK3 at both the mRNA and protein levels using RT-PCR and Western blotting. RESULTS In public datasets and clinical samples, ELK3 was highly expressed in gastric cancer (P < 0.05), and its expression increased with the progression of M stage, AJCC stage, and perineural invasion (P < 0.05). ELK3 expression was correlated with N stage, AJCC stage, Lauren classification, differentiation, pathological classification, and microsatellite status of gastric cancer (P < 0.05). A high expression of ELK3 was associated with significantly reduced overall survival and disease-free survival of the patients, and served as an independent prognostic factor of gastric cancer (P < 0.05). Comprehensive analysis identified 176 potential target genes of ELK3, and enrichment analysis showed that ELK3 may regulate Rap1, AMPK, chemokines, VEGF, TNF, and tumor PD-L1/PD-1 signaling (PP < 0.05). The expression of ELK3 was negatively correlated with regulatory T cells, follicular helper T cells, and CD8+T cells in gastric cancer (P < 0.05). CONCLUSION ELK3 acts as an oncogene in gastric cancer, and its high expression may promote the occurrence, progression and immune escape of gastric cancer.
Collapse
Affiliation(s)
- 立强 周
- />南昌大学第二附属医院胃肠外科,江西 南昌 330006Department of Gastrointestinal Surgery, Second Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - 忧 吴
- />南昌大学第二附属医院胃肠外科,江西 南昌 330006Department of Gastrointestinal Surgery, Second Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - 林 辛
- />南昌大学第二附属医院胃肠外科,江西 南昌 330006Department of Gastrointestinal Surgery, Second Affiliated Hospital of Nanchang University, Nanchang 330006, China
| |
Collapse
|
24
|
Baj J, Forma A, Flieger W, Morawska I, Michalski A, Buszewicz G, Sitarz E, Portincasa P, Garruti G, Flieger M, Teresiński G. Helicobacter pylori Infection and Extragastric Diseases-A Focus on the Central Nervous System. Cells 2021; 10:2191. [PMID: 34571840 PMCID: PMC8469861 DOI: 10.3390/cells10092191] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 08/22/2021] [Accepted: 08/23/2021] [Indexed: 02/07/2023] Open
Abstract
Helicobacter pylori (H. pylori) is most known to cause a wide spectrum of gastrointestinal impairments; however, an increasing number of studies indicates that H. pylori infection might be involved in numerous extragastric diseases such as neurological, dermatological, hematologic, ocular, cardiovascular, metabolic, hepatobiliary, or even allergic diseases. In this review, we focused on the nervous system and aimed to summarize the findings regarding H. pylori infection and its involvement in the induction/progression of neurological disorders. Neurological impairments induced by H. pylori infection are primarily due to impairments in the gut-brain axis (GBA) and to an altered gut microbiota facilitated by H. pylori colonization. Currently, regarding a potential relationship between Helicobacter infection and neurological disorders, most of the studies are mainly focused on H. pylori.
Collapse
Affiliation(s)
- Jacek Baj
- Chair and Department of Anatomy, Medical University of Lublin, Jaczewskiego 4, 20-090 Lublin, Poland;
| | - Alicja Forma
- Department of Forensic Medicine, Medical University of Lublin, 20-090 Lublin, Poland; (A.F.); (G.B.); (M.F.); (G.T.)
| | - Wojciech Flieger
- Chair and Department of Anatomy, Medical University of Lublin, Jaczewskiego 4, 20-090 Lublin, Poland;
| | - Izabela Morawska
- Department of Clinical Immunology and Immunotherapy, Medical University of Lublin, 20-093 Lublin, Poland; (I.M.); (A.M.)
| | - Adam Michalski
- Department of Clinical Immunology and Immunotherapy, Medical University of Lublin, 20-093 Lublin, Poland; (I.M.); (A.M.)
| | - Grzegorz Buszewicz
- Department of Forensic Medicine, Medical University of Lublin, 20-090 Lublin, Poland; (A.F.); (G.B.); (M.F.); (G.T.)
| | - Elżbieta Sitarz
- Chair and I Department of Psychiatry, Psychotherapy, and Early Intervention, Medical University of Lublin, 20-439 Lublin, Poland;
| | - Piero Portincasa
- Clinica Medica “A. Murri”, Department of Biomedical Sciences & Human Oncology, University of Bari Medical School, 70124 Bari, Italy;
| | - Gabriella Garruti
- Section of Endocrinology, Department of Emergency and Organ Transplantations, University of Bari “Aldo Moro” Medical School, Piazza G. Cesare 11, 70124 Bari, Italy;
| | - Michał Flieger
- Department of Forensic Medicine, Medical University of Lublin, 20-090 Lublin, Poland; (A.F.); (G.B.); (M.F.); (G.T.)
| | - Grzegorz Teresiński
- Department of Forensic Medicine, Medical University of Lublin, 20-090 Lublin, Poland; (A.F.); (G.B.); (M.F.); (G.T.)
| |
Collapse
|
25
|
Rihawi K, Ricci AD, Rizzo A, Brocchi S, Marasco G, Pastore LV, Llimpe FLR, Golfieri R, Renzulli M. Tumor-Associated Macrophages and Inflammatory Microenvironment in Gastric Cancer: Novel Translational Implications. Int J Mol Sci 2021; 22:3805. [PMID: 33916915 PMCID: PMC8067563 DOI: 10.3390/ijms22083805] [Citation(s) in RCA: 107] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 03/30/2021] [Accepted: 04/05/2021] [Indexed: 02/07/2023] Open
Abstract
Gastric cancer (GC) represents the fifth most frequently diagnosed cancer worldwide, with a poor prognosis in patients with advanced disease despite many improvements in systemic treatments in the last decade. In fact, GC has shown resistance to several treatment options, and thus, notable efforts have been focused on the research and identification of novel therapeutic targets in this setting. The tumor microenvironment (TME) has emerged as a potential therapeutic target in several malignancies including GC, due to its pivotal role in cancer progression and drug resistance. Therefore, several agents and therapeutic strategies targeting the TME are currently under assessment in both preclinical and clinical studies. The present study provides an overview of available evidence of the inflammatory TME in GC, highlighting different types of tumor-associated cells and implications for future therapeutic strategies.
Collapse
Affiliation(s)
- Karim Rihawi
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (K.R.); (A.D.R.); (A.R.); (F.L.R.L.)
| | - Angela Dalia Ricci
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (K.R.); (A.D.R.); (A.R.); (F.L.R.L.)
| | - Alessandro Rizzo
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (K.R.); (A.D.R.); (A.R.); (F.L.R.L.)
| | - Stefano Brocchi
- Department of Radiology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (S.B.); (L.V.P.); (R.G.)
| | - Giovanni Marasco
- Department of Medical and Surgical Sciences, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy;
| | - Luigi Vincenzo Pastore
- Department of Radiology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (S.B.); (L.V.P.); (R.G.)
| | - Fabiola Lorena Rojas Llimpe
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (K.R.); (A.D.R.); (A.R.); (F.L.R.L.)
| | - Rita Golfieri
- Department of Radiology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (S.B.); (L.V.P.); (R.G.)
| | - Matteo Renzulli
- Department of Radiology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (S.B.); (L.V.P.); (R.G.)
| |
Collapse
|
26
|
Forma A, Chilimoniuk Z, Januszewski J, Sitarz R. The Potential Application of Allium Extracts in the Treatment of Gastrointestinal Cancers. GASTROENTEROLOGY INSIGHTS 2021; 12:136-146. [DOI: 10.3390/gastroent12020012] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Herbal medicine is currently widely practiced, since natural resources are reported to alleviate side effects during oncological treatment while modifying cancer cell responses at the same time. Allium vegetables and their constituents have recently been extensively investigated due to the numerous possible beneficial properties, establishing them as an additional treatment modality in different cancers. According to the epidemiological evidence, as well as many in vivo and in vitro studies, the abovementioned substances seem to be effective in the prevention and inhibition of the progression of carcinogenesis. Due to high concentrations of organosulfur compounds, which exhibit anticarcinogenic, antimicrobial, as well as anti-inflammatory properties, Allium constituents are believed to constitute a promising prevention and supportive therapy for oncological patients. Besides, it was demonstrated that a combination of Allium extracts with chemotherapy provided satisfactory clinical outcomes while at the same time being cost-effective. The aim of this review was to present and discuss currently investigated Allium extracts and their effects on several gastrointestinal cancers including gastric, colon, liver, esophageal, and pancreatic cancer.
Collapse
Affiliation(s)
- Alicja Forma
- Department of Forensic Medicine, Medical University of Lublin, 20-090 Lublin, Poland
| | - Zuzanna Chilimoniuk
- Department of Forensic Medicine, Medical University of Lublin, 20-090 Lublin, Poland
| | - Jacek Januszewski
- Department of Forensic Medicine, Medical University of Lublin, 20-090 Lublin, Poland
| | - Robert Sitarz
- Department of Human Anatomy, Medical University of Lublin, 20-090 Lublin, Poland
- Department of Surgical Oncology, Center of Oncology of the Lublin Region St. Jana z Dukli, 20-091 Lublin, Poland
| |
Collapse
|
27
|
Tyczyńska M, Kędzierawski P, Karakuła K, Januszewski J, Kozak K, Sitarz M, Forma A. Treatment Strategies of Gastric Cancer-Molecular Targets for Anti-angiogenic Therapy: a State-of-the-art Review. J Gastrointest Cancer 2021; 52:476-488. [PMID: 33761051 PMCID: PMC8131337 DOI: 10.1007/s12029-021-00629-7] [Citation(s) in RCA: 3] [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] [Accepted: 03/14/2021] [Indexed: 12/19/2022]
Abstract
Purpose Recent studies have suggested that molecular targets for the anti-angiogenic therapy might constitute a basis for additional therapy in gastric cancer treatment. A vast number of molecules, receptors, pathways, specific interactions, and thus strategies that target gastric cancer angiogenesis specifically have been reported in numerous research articles and clinical trials. Methods We conducted a systematic literature review of molecularly targeted treatment strategies in gastric cancer on the following databases—PubMed, Google Scholar, and Scopus—on September 20, 2020. Multiple articles and evaluations were searched for studies reporting newly found and promising molecular anti-angiogenic therapy pathways. Eventually, 39 articles regarding the anti-angiogenic therapy in gastric cancer were included in the final analysis. Results As a consequence of the release of the pro-angiogenic molecules from the tumour cells, gastric cancer presents high angiogenic capability. Therefore, potential schemes for future treatment strategies include the decrease of the process ligands as well as the expression of their receptors. Moreover, the increase in the angiogenic inhibitor levels and direct aim for the inner walls of the endothelial cells appear as a promising therapeutic strategy. Beyond that, angiogenesis process inhibition seems to indirectly exaggerate the effects of chemotherapy in the considered patients. Conclusions The anti-angiogenic treatment in gastric cancer patients evaluates its significance especially in the early stages of the malignancy. The studies conducted so far show that most of the meaningful angiogenic factors and receptors with the potential molecular pathways should be further evaluated since they could potentially play a substantial role in future therapies.
Collapse
Affiliation(s)
- Magdalena Tyczyńska
- Department of Human Anatomy, Medical University of Lublin, 20-090 Lublin, Poland
| | - Paweł Kędzierawski
- Department of Forensic Medicine, Medical University of Lublin, 20-090 Lublin, Poland
| | - Kaja Karakuła
- Department of Psychiatry, Psychotherapy and Early Intervention, Medical University of Lublin, Gluska Street 1, 20-439 Lublin, Poland
| | - Jacek Januszewski
- Department of Forensic Medicine, Medical University of Lublin, 20-090 Lublin, Poland
| | - Krzysztof Kozak
- Department of Human Anatomy, Medical University of Lublin, 20-090 Lublin, Poland
| | - Monika Sitarz
- Department of Conservative Dentistry with Endodontics, Medical University of Lublin, 20-090 Lublin, Poland
| | - Alicja Forma
- Department of Forensic Medicine, Medical University of Lublin, 20-090 Lublin, Poland
| |
Collapse
|
28
|
Ganoderma lucidum Spore Polysaccharide Inhibits the Growth of Hepatocellular Carcinoma Cells by Altering Macrophage Polarity and Induction of Apoptosis. J Immunol Res 2021; 2021:6696606. [PMID: 33748291 PMCID: PMC7954632 DOI: 10.1155/2021/6696606] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 02/02/2021] [Accepted: 02/10/2021] [Indexed: 12/14/2022] Open
Abstract
Background Ganoderma lucidum has certain components with known pharmacological effects, including strengthening immunity and anti-inflammatory activity. G. lucidum seeds inherit all its biological characteristics. G. lucidum spore polysaccharide (GLSP) is the main active ingredient to enhance these effects. However, its specific biological mechanisms are not exact. Our research is aimed at revealing the specific biological mechanism of GLSP to enhance immunity and inhibit the growth of H22 hepatocellular carcinoma cells. Methods We extracted primary macrophages (Mø) from BALB/c mice and treated them with GLSP (800 μg/mL, 400 μg/mL, and 200 μg/mL) to observe its effects on macrophage polarization and cytokine secretion. We used GLSP and GLSP-intervened macrophage supernatant to treat H22 tumor cells and observed their effects using MTT and flow cytometry. Moreover, real-time fluorescent quantitative PCR and western blotting were used to observe the effect of GLSP-intervened macrophage supernatant on the PI3K/AKT and mitochondrial apoptosis pathways. Results In this study, GLSP promoted the polarization of primary macrophages to M1 type and the upregulation of some cytokines such as TNF-α, IL-1β, IL-6, and TGF-β1. The MTT assay revealed that GLSP+Mø at 400 μg/mL and 800 μg/mL significantly inhibited H22 cell proliferation in a dose-dependent manner. Flow cytometry analysis revealed that GLSP+Mø induced apoptosis and cell cycle arrest at the G2/M phase, associated with the expression of critical genes and proteins (PI3K, p-AKT, BCL-2, BAX, and caspase-9) that regulate the PI3K/AKT pathway and apoptosis. GLSP reshapes the tumor microenvironment by activating macrophages, promotes the polarization of primary macrophages to M1 type, and promotes the secretion of various inflammatory factors and cytokines. Conclusion Therefore, as a natural nutrient, GLSP is a potential agent in hepatocellular carcinoma cell treatment and induction of apoptosis.
Collapse
|