1
|
Liu CY, Li Z, Cheng FE, Nan Y, Li WQ. Radix Codonopsis: a review of anticancer pharmacological activities. Front Pharmacol 2025; 15:1498707. [PMID: 39840099 PMCID: PMC11747557 DOI: 10.3389/fphar.2024.1498707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Accepted: 12/12/2024] [Indexed: 01/23/2025] Open
Abstract
Radix Codonopsis (Dangshen), derived from the dried root of plants in the Campanulaceae family, is a widely used Chinese herbal medicine. It is renowned for its pharmacological effects, including tonifying the middle qi, invigorating the spleen, benefiting the lungs, enhancing immunity, and nourishing the blood. Codonopsis extract is frequently incorporated into health products such as tablets and capsules, making it accessible for daily health maintenance. Additionally, it is commonly used in dietary applications like soups, teas, and porridges to nourish qi, enrich blood, and promote overall vitality. In recent years, increasing attention has been given to the anti-cancer potential of Radix Codonopsis. Studies have identified key active components such as luteolin, stigmasterol, polyacetylenes, lobetyolin, and glycitein, which exhibit anti-tumor properties through mechanisms like inhibiting cancer cell growth and proliferation, suppressing epithelial-mesenchymal transition (EMT), and inducing apoptosis. This review highlights the research progress on Radix Codonopsis, including its active constituents, anti-cancer mechanisms, and its role in the convergence of medicine and food in modern life. By doing so, it aims to provide valuable insights and references for future scientific studies and clinical applications of Radix Codonopsis.
Collapse
Affiliation(s)
- Cai-Yue Liu
- Ningxia Medical University, Ningxia of Traditional Chinese Medicine, Yinchuan, China
| | - Zheng Li
- Ningxia Medical University, Ningxia of Traditional Chinese Medicine, Yinchuan, China
| | - Fan-E. Cheng
- Ningxia Medical University, Ningxia of Traditional Chinese Medicine, Yinchuan, China
| | - Yi Nan
- Ningxia Medical University, Ningxia of Traditional Chinese Medicine, Yinchuan, China
- Key Laboratory of Ningxia Minority Medicine Modernization, Ministry of Education, Ningxia Medical University, Yinchuan, China
| | - Wei-Qiang Li
- Ningxia Medical University, Ningxia of Traditional Chinese Medicine, Yinchuan, China
- Key Laboratory of Ningxia Minority Medicine Modernization, Ministry of Education, Ningxia Medical University, Yinchuan, China
| |
Collapse
|
2
|
Salazar-Saura I, Pinilla-Sala M, Megías J, Navarro L, Roselló-Sastre E, San-Miguel T. Pericytes in Glioblastoma: Hidden Regulators of Tumor Vasculature and Therapy Resistance. Cancers (Basel) 2024; 17:15. [PMID: 39796646 PMCID: PMC11718950 DOI: 10.3390/cancers17010015] [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: 11/27/2024] [Revised: 12/19/2024] [Accepted: 12/22/2024] [Indexed: 01/13/2025] Open
Abstract
Glioblastoma IDH wild type (GB), the most common malignant primary brain tumor, is characterized by rapid proliferation, extensive infiltration into surrounding brain tissue, and significant resistance to current therapies. Median survival is only 15 months despite extensive clinical efforts. The tumor microenvironment (TME) in GB is highly specialized, supporting the tumor's aggressive behavior and its ability to evade conventional treatments. One critical component is the aberrant vascular network that complicates the delivery of chemotherapy across the blood-brain barrier. Antiangiogenic therapies emerged as a promising option but have shown limited efficacy in extending the survival of these patients. Comprehension of the complex vascular network of GB may be a key to overcoming the limitations of current therapies. Pericytes are gaining recognition within the context of the TME. These mural cells are essential for vascular integrity and may contribute to tumor progression and therapeutic resistance. Although their role has been evidenced in other tumors, they remain underexplored in GB. Pericytes are known to respond to tumor hypoxia and interact with vascular endothelia, influencing responses to DNA damage and antiangiogenic treatments. They actively regulate not only angiogenesis but also the different vasculogenic strategies for tumor neovascularization. Additionally, they affect leukocyte trafficking and tumor-associated macrophages. This review aims to integrate the various functions controlled by pericytes to favor deeper investigation into their actionable potential. Pericytes may represent a promising target for novel therapeutic strategies in order to improve patient outcomes.
Collapse
Affiliation(s)
- Irene Salazar-Saura
- Pathology Service, Consorcio Hospital General Universitario de Valencia, 46014 Valencia, Spain; (I.S.-S.); (L.N.); (E.R.-S.)
| | - María Pinilla-Sala
- Research Group on Tumors of the Central Nervous System, Pathology Department, University of Valencia, 46010 Valencia, Spain;
- INCLIVA Foundation, 46010 Valencia, Spain
| | - Javier Megías
- Research Group on Tumors of the Central Nervous System, Pathology Department, University of Valencia, 46010 Valencia, Spain;
- INCLIVA Foundation, 46010 Valencia, Spain
| | - Lara Navarro
- Pathology Service, Consorcio Hospital General Universitario de Valencia, 46014 Valencia, Spain; (I.S.-S.); (L.N.); (E.R.-S.)
| | - Esther Roselló-Sastre
- Pathology Service, Consorcio Hospital General Universitario de Valencia, 46014 Valencia, Spain; (I.S.-S.); (L.N.); (E.R.-S.)
| | - Teresa San-Miguel
- Research Group on Tumors of the Central Nervous System, Pathology Department, University of Valencia, 46010 Valencia, Spain;
- INCLIVA Foundation, 46010 Valencia, Spain
| |
Collapse
|
3
|
Greaves GE, Pinna A, Taylor JM, Porter AE, Phillips CC. In Depth Mapping of Mesoporous Silica Nanoparticles in Malignant Glioma Cells Using Scattering-Type Scanning Near-Field Optical Microscopy. CHEMICAL & BIOMEDICAL IMAGING 2024; 2:842-849. [PMID: 39735833 PMCID: PMC11672216 DOI: 10.1021/cbmi.4c00053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 10/09/2024] [Accepted: 10/10/2024] [Indexed: 12/31/2024]
Abstract
Mesoporous silica nanoparticles (MSNPs) are promising nanomedicine vehicles due to their biocompatibility and ability to carry large cargoes. It is critical in nanomedicine development to be able to map their uptake in cells, including distinguishing surface associated MSNPs from those that are embedded or internalized into cells. Conventional nanoscale imaging techniques, such as electron and fluorescence microscopies, however, generally require the use of stains and labels to image both the biological material and the nanomedicines, which can interfere with the biological processes at play. We demonstrate an alternative imaging technique for investigating the interactions between cells and nanostructures, scattering-type scanning near-field optical microscopy (s-SNOM). s-SNOM combines the chemical sensitivity of infrared spectroscopy with the nanoscale spatial resolving power of scanning probe microscopy. We use the technique to chemically map the uptake of MSNPs in whole human glioblastoma cells and show that the simultaneously acquired topographical information can provide the embedding status of the MSNPs. We focus our imaging efforts on the lamellipodia and filopodia structures at the peripheries of the cells due to their significance in cancer invasiveness.
Collapse
Affiliation(s)
- George E. Greaves
- Experimental
Solid State Physics Group, Department of Physics, Imperial College, Exhibition Road, SW72AZ London, U.K.
| | - Alessandra Pinna
- Department
of Materials and London Centre for Nanotechnology, Imperial College, Exhibition
Road, SW72AZ London, U.K.
- School
of Veterinary Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford GU2 7XH, U.K.
- The
Francis Crick Institute, NW1 1AT London, U.K.
| | - Jonathan M. Taylor
- Department
of Materials and London Centre for Nanotechnology, Imperial College, Exhibition
Road, SW72AZ London, U.K.
| | - Alexandra E. Porter
- Department
of Materials and London Centre for Nanotechnology, Imperial College, Exhibition
Road, SW72AZ London, U.K.
| | - Chris C. Phillips
- Experimental
Solid State Physics Group, Department of Physics, Imperial College, Exhibition Road, SW72AZ London, U.K.
| |
Collapse
|
4
|
Alexopoulos S, McGawley M, Mathews R, Papakostopoulou S, Koulas S, Leonidas DD, Zwain T, Hayes JM, Skamnaki V. Evidence for the Quercetin Binding Site of Glycogen Phosphorylase as a Target for Liver-Isoform-Selective Inhibitors against Glioblastoma: Investigation of Flavanols Epigallocatechin Gallate and Epigallocatechin. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:24070-24081. [PMID: 39433280 PMCID: PMC11528470 DOI: 10.1021/acs.jafc.4c06920] [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: 08/01/2024] [Revised: 10/07/2024] [Accepted: 10/08/2024] [Indexed: 10/23/2024]
Abstract
Glycogen phosphorylase (GP) is the rate-determining enzyme in glycogenolysis, and its druggability has been extensively studied over the years for the development of therapeutics against type 2 diabetes (T2D) and, more recently, cancer. However, the conservation of binding sites between the liver and muscle isoforms makes the inhibitor selectivity challenging. Using a combination of kinetic, crystallographic, modeling, and cellular studies, we have probed the binding of dietary flavonoids epigallocatechin gallate (EGCG) and epigallocatechin (EGC) to GP isoforms. The structures of rmGPb-EGCG and rmGPb-EGC complexes were determined by X-ray crystallography, showing binding at the quercetin binding site (QBS) in agreement with kinetic studies that revealed both compounds as noncompetitive inhibitors of GP, with EGCG also causing a significant reduction in cell viability and migration of U87-MG glioblastoma cells. Interestingly, EGCG exhibits different binding modes to GP isoforms, revealing QBS as a promising site for GP targeting, offering new opportunities for the design of liver-selective GP inhibitors.
Collapse
Affiliation(s)
- Serafeim Alexopoulos
- Department
of Biochemistry and Biotechnology, University
of Thessaly, Biopolis, Larisa 41500, Greece
| | - Megan McGawley
- School
of Pharmacy & Biomedical Sciences, University
of Central Lancashire, Preston PR1 2HE, U.K.
| | - Roshini Mathews
- School
of Pharmacy & Biomedical Sciences, University
of Central Lancashire, Preston PR1 2HE, U.K.
| | - Souzana Papakostopoulou
- Department
of Biochemistry and Biotechnology, University
of Thessaly, Biopolis, Larisa 41500, Greece
| | - Symeon Koulas
- Department
of Biochemistry and Biotechnology, University
of Thessaly, Biopolis, Larisa 41500, Greece
| | - Demetres D. Leonidas
- Department
of Biochemistry and Biotechnology, University
of Thessaly, Biopolis, Larisa 41500, Greece
| | - Tamara Zwain
- School
of Pharmacy & Biomedical Sciences, University
of Central Lancashire, Preston PR1 2HE, U.K.
| | - Joseph M. Hayes
- School
of Pharmacy & Biomedical Sciences, University
of Central Lancashire, Preston PR1 2HE, U.K.
| | - Vasiliki Skamnaki
- Department
of Biochemistry and Biotechnology, University
of Thessaly, Biopolis, Larisa 41500, Greece
| |
Collapse
|
5
|
Tsai WE, Liu YT, Kuo FH, Cheng WY, Shen CC, Chiao MT, Huang YF, Liang YJ, Yang YC, Hsieh WY, Chen JP, Liu SY, Chiu CD. Crocetin Enhances Temozolomide Efficacy in Glioblastoma Therapy Through Multiple Pathway Suppression. Curr Neurovasc Res 2024; 21:320-336. [PMID: 39092730 DOI: 10.2174/0115672026332275240731054001] [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: 05/23/2024] [Revised: 06/25/2024] [Accepted: 06/28/2024] [Indexed: 08/04/2024]
Abstract
BACKGROUND Glioblastoma multiforme (GBM) is an aggressive type of brain tumor that is difficult to remove surgically. Research suggests that substances from saffron, namely crocetin and crocin, could be effective natural treatments, showing abilities to kill cancer cells. METHODS Our study focused on evaluating the effects of crocetin on glioma using the U87 cell line. We specifically investigated how crocetin affects the survival, growth, and spread of glioma cells, exploring its impact at concentrations ranging from 75-150 μM. The study also included experiments combining crocetin with the chemotherapy drug Temozolomide (TMZ) to assess potential synergistic effects. RESULTS Crocetin significantly reduced the viability, proliferation, and migration of glioma cells. It achieved these effects by decreasing the levels of Matrix Metallopeptidase 9 (MMP-9) and Ras homolog family member A (RhoA), proteins that are critical for cancer progression. Additionally, crocetin inhibited the formation of cellular structures necessary for tumor growth. It blocked multiple points of the Ak Strain Transforming (AKT) signaling pathway, which is vital for cancer cell survival. This treatment led to increased cell death and disrupted the cell cycle in the glioma cells. When used in combination with TMZ, crocetin not only enhanced the reduction of cancer cell growth but also promoted cell death and reduced cell replication. This combination therapy further decreased levels of high mobility group box 1 (HMGB1) and Receptor for Advanced Glycation End-products (RAGE), proteins linked to inflammation and tumor progression. It selectively inhibited certain pathways involved in the cellular stress response without affecting others. CONCLUSION Our results underscore the potential of crocetin as a treatment for glioma. It targets various mechanisms involved in tumor growth and spread, offering multiple avenues for therapy. Further studies are essential to fully understand and utilize crocetin's benefits in treating glioma.
Collapse
Affiliation(s)
- Wei-En Tsai
- Taichung Municipal Taichung First Senior High School, Taichung, Taiwan
| | - Yen-Tsen Liu
- Taichung Municipal Taichung First Senior High School, Taichung, Taiwan
| | - Fu-Hsuan Kuo
- Center for Geriatrics and Gerontology, Taichung Veterans Hospital, Taichung, 40705, Taiwan
| | - Wen-Yu Cheng
- Department of Minimally Invasive Skull Base Neurosurgery, Neurological Institute, Taichung Veterans General Hospital, Taichung, Taiwan
- Department of Physical Therapy, Hung Kuang University, Taichung, Taiwan
- Institute of Biomedical Sciences, National Chung Hsing University, Taichung, Taiwan
- Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing University, Taichung, Taiwan
| | - Chiung-Chyi Shen
- Department of Minimally Invasive Skull Base Neurosurgery, Neurological Institute, Taichung Veterans General Hospital, Taichung, Taiwan
- Department of Physical Therapy, Hung Kuang University, Taichung, Taiwan
- Basic Medical Education, Central Taiwan University of Science and Technology, Taichung, Taiwan
| | - Ming-Tsang Chiao
- Department of Minimally Invasive Skull Base Neurosurgery, Neurological Institute, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Yu-Fen Huang
- Department of Minimally Invasive Skull Base Neurosurgery, Neurological Institute, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Yea-Jiuen Liang
- Department of Minimally Invasive Skull Base Neurosurgery, Neurological Institute, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Yi-Chin Yang
- Department of Minimally Invasive Skull Base Neurosurgery, Neurological Institute, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Wan-Yu Hsieh
- Department of Minimally Invasive Skull Base Neurosurgery, Neurological Institute, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Jun-Peng Chen
- Biostatistics Task Force, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Szu-Yuan Liu
- Department of Neurosurgery, Oncology Neurosurgery Division, Neurological Institute, Taichung Veterans General Hospital, Taichung, Taiwan
- Department of Life Science, College of Life Science, Graduate Institute of Life Science, National Chung Hsing University, Taichung, Taiwan
| | - Cheng-Di Chiu
- Spine Center, China Medical University Hospital, Taichung, Taiwan
- Department of Neurosurgery, China Medical University Hospital, Taichung, Taiwan
| |
Collapse
|
6
|
Macedo C, Costa PC, Rodrigues F. Bioactive compounds from Actinidia arguta fruit as a new strategy to fight glioblastoma. Food Res Int 2024; 175:113770. [PMID: 38129059 DOI: 10.1016/j.foodres.2023.113770] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 11/10/2023] [Accepted: 11/22/2023] [Indexed: 12/23/2023]
Abstract
In recent years, there has been a significant demand for natural products as a mean of disease prevention or as an alternative to conventional medications. The driving force for this change is the growing recognition of the abundant presence of valuable bioactive compounds in natural products. On recent years Actinia arguta fruit, also known as kiwiberry, has attracted a lot of attention from scientific community due to its richness in bioactive compounds, including phenolic compounds, organic acids, vitamins, carotenoids and fiber. These bioactive compounds contribute to the fruit's diverse outstanding biological activities such as antioxidant, anti-inflammatory, neuroprotective, immunomodulatory, and anti-cancer properties. Due to these properties, the fruit may have the potential to be used in the treatment/prevention of various types of cancer, including glioblastoma. Glioblastoma is the most aggressive form of brain cancer, displaying 90 % of recurrence rate within a span of 2 years. Despite the employment of an aggressive approach, the prognosis remains unfavorable, emphasizing the urgent requirement for the development of new effective treatments. The preclinical evidence suggests that kiwiberry has potential impact on glioblastoma by reducing the cancer self-renewal, modulating the signaling pathways involved in the regulation of the cell phenotype and metabolism, and influencing the consolidation of the tumor microenvironment. Even though, challenges such as the imprecise composition and concentration of bioactive compounds, and its low bioavailability after oral administration may be drawbacks to the development of kiwiberry-based treatments, being urgent to ensure the safety and efficacy of kiwiberry for the prevention and treatment of glioblastoma. This review aims to highlight the potential impact of A. arguta bioactive compounds on glioblastoma, providing novel insights into their applicability as complementary or alternative therapies.
Collapse
Affiliation(s)
- Catarina Macedo
- REQUIMTE/LAQV, ISEP, Polytechnic of Porto, Rua Dr. António Bernardino de Almeida, 4249-015 Porto, Portugal; REQUIMTE/UCIBIO, MedTech-Laboratory of Pharmaceutical Technology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal; Associate Laboratory i4HB-Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Paulo C Costa
- REQUIMTE/UCIBIO, MedTech-Laboratory of Pharmaceutical Technology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal; Associate Laboratory i4HB-Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal.
| | - Francisca Rodrigues
- REQUIMTE/LAQV, ISEP, Polytechnic of Porto, Rua Dr. António Bernardino de Almeida, 4249-015 Porto, Portugal.
| |
Collapse
|
7
|
Therapeutic Potential of Luteolin on Cancer. Vaccines (Basel) 2023; 11:vaccines11030554. [PMID: 36992138 DOI: 10.3390/vaccines11030554] [Citation(s) in RCA: 47] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 02/23/2023] [Accepted: 02/24/2023] [Indexed: 03/03/2023] Open
Abstract
Cancer is a global concern, as the rate of incidence is increasing each year. The challenges related to the current chemotherapy drugs, such as the concerns related to toxicity, turn to cancer therapeutic research to discover alternative therapy strategies that are less toxic to normal cells. Among those studies, the use of flavonoids—natural compounds produced by plants as secondary metabolites for cancer therapy—has been a hot topic in cancer treatment. Luteolin, a flavonoid that has been present in many fruits, vegetables, and herbs, has been identified to exhibit numerous biological activities, including anti-inflammatory, antidiabetic, and anticancer properties. The anticancer property of Luteolin has been extensively researched in many cancer types and has been related to its ability to inhibit tumor growth by targeting cellular processes such as apoptosis, angiogenesis, migration, and cell cycle progression. It achieves this by interacting with various signaling pathways and proteins. In the current review, the molecular targets of Luteolin as it exerts its anticancer properties, the combination therapy that includes Luteolin with other flavonoids or chemotherapeutic drugs, and the nanodelivery strategies for Luteolin are described for several cancer types.
Collapse
|
8
|
Progress in targeting PTEN/PI3K/Akt axis in glioblastoma therapy: Revisiting molecular interactions. Biomed Pharmacother 2023; 158:114204. [PMID: 36916430 DOI: 10.1016/j.biopha.2022.114204] [Citation(s) in RCA: 61] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 12/16/2022] [Accepted: 12/30/2022] [Indexed: 01/05/2023] Open
Abstract
Glioblastoma (GBM) is one of the most malignant cancers of central nervous system and due to its sensitive location, surgical resection has high risk and therefore, chemotherapy and radiotherapy are utilized for its treatment. However, chemoresistance and radio-resistance are other problems in GBM treatment. Hence, new therapies based on genes are recommended for treatment of GBM. PTEN is a tumor-suppressor operator in cancer that inhibits PI3K/Akt/mTOR axis in diminishing growth, metastasis and drug resistance. In the current review, the function of PTEN/PI3K/Akt axis in GBM progression is evaluated. Mutation or depletion of PTEN leads to increase in GBM progression. Low expression level of PTEN mediates poor prognosis in GBM and by increasing proliferation and invasion, promotes malignancy of tumor cells. Moreover, loss of PTEN signaling can result in therapy resistance in GBM. Activation of PTEN signaling impairs GBM metabolism via glycolysis inhibition. In contrast to PTEN, PI3K/Akt signaling has oncogenic function and during tumor progression, expression level of PI3K/Akt enhances. PI3K/Akt signaling shows positive association with oncogenic pathways and its expression similar to PTEN signaling, is regulated by non-coding RNAs. PTEN upregulation and PI3K/Akt signaling inhibition by anti-cancer agents can be beneficial in interfering GBM progression. This review emphasizes on the signaling networks related to PTEN/PI3K/Akt and provides new insights for targeting this axis in effective GBM treatment.
Collapse
|
9
|
Chen YH, Wu JX, Yang SF, Hsiao YH. Synergistic Combination of Luteolin and Asiatic Acid on Cervical Cancer In Vitro and In Vivo. Cancers (Basel) 2023; 15:cancers15020548. [PMID: 36672499 PMCID: PMC9857275 DOI: 10.3390/cancers15020548] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 01/11/2023] [Accepted: 01/13/2023] [Indexed: 01/18/2023] Open
Abstract
Cervical cancer is an important issue globally because it is the second most common gynecological malignant tumor and conventional treatment effects have been shown to be limited. Lut and AsA are plant-derived natural flavonoid and triterpenoid products that have exhibited anticancer activities and can modulate various signaling pathways. Thus, the aim of the present study was to evaluate whether Lut combined with AsA could enhance the anticancer effect to inhibit cervical cancer cell proliferation and examine the underlying molecular mechanisms in vitro and in vivo. The results of a CCK-8 assay showed that Lut combined with AsA more effectively inhibited the proliferation of CaSki and HeLa cells than Lut or AsA treatment alone. Lut combined with AsA caused apoptosis induction and sub-G1-phase arrest in CaSki and HeLa cells, as confirmed by flow cytometry, mitoROS analysis, antioxidant activity measurement and western blot assay. In addition, Lut combined with AsA significantly inhibited the cell migration ability of CaSki and HeLa cells in a wound-healing assay. Furthermore, Lut combined with AsA induced apoptosis and inhibited migration through downregulated PI3K/AKT (PI3K, AKT and p70S6K), JNK/p38 MAPK and FAK (integrin β1, paxillin and FAK) signaling and upregulated ERK signaling. In an in vivo study, Lut combined with AsA markedly inhibited cervical cancer cell-derived xenograft tumor growth. Collectively, the present study showed that Lut combined with AsA may be used as an anticancer agent to improve the prognosis of cervical cancer. Indeed, with additional research to develop standardized dosages, Lut and AsA combination therapy could also be applied in clinical medicine.
Collapse
Affiliation(s)
- Ya-Hui Chen
- Women’s Health Research Laboratory, Changhua Christian Hospital, Changhua 50006, Taiwan
- Institute of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan
| | - Jyun-Xue Wu
- Women’s Health Research Laboratory, Changhua Christian Hospital, Changhua 50006, Taiwan
| | - Shun-Fa Yang
- Institute of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung 40201, Taiwan
| | - Yi-Hsuan Hsiao
- Women’s Health Research Laboratory, Changhua Christian Hospital, Changhua 50006, Taiwan
- Institute of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan
- Department of Obstetrics and Gynecology, Changhua Christian Hospital, Changhua 50006, Taiwan
- School of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan
- College of Medicine, Kaohsiung Medical University, Kaohsiung 807378, Taiwan
- College of Medicine, National Chung Hsing University, Taichung 40227, Taiwan
- Correspondence:
| |
Collapse
|
10
|
Mitra S, Dash R, Munni YA, Selsi NJ, Akter N, Uddin MN, Mazumder K, Moon IS. Natural Products Targeting Hsp90 for a Concurrent Strategy in Glioblastoma and Neurodegeneration. Metabolites 2022; 12:1153. [PMID: 36422293 PMCID: PMC9697676 DOI: 10.3390/metabo12111153] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 11/16/2022] [Accepted: 11/16/2022] [Indexed: 09/16/2023] Open
Abstract
Glioblastoma multiforme (GBM) is one of the most common aggressive, resistant, and invasive primary brain tumors that share neurodegenerative actions, resembling many neurodegenerative diseases. Although multiple conventional approaches, including chemoradiation, are more frequent in GBM therapy, these approaches are ineffective in extending the mean survival rate and are associated with various side effects, including neurodegeneration. This review proposes an alternative strategy for managing GBM and neurodegeneration by targeting heat shock protein 90 (Hsp90). Hsp90 is a well-known molecular chaperone that plays essential roles in maintaining and stabilizing protein folding to degradation in protein homeostasis and modulates signaling in cancer and neurodegeneration by regulating many client protein substrates. The therapeutic benefits of Hsp90 inhibition are well-known for several malignancies, and recent evidence highlights that Hsp90 inhibitors potentially inhibit the aggressiveness of GBM, increasing the sensitivity of conventional treatment and providing neuroprotection in various neurodegenerative diseases. Herein, the overview of Hsp90 modulation in GBM and neurodegeneration progress has been discussed with a summary of recent outcomes on Hsp90 inhibition in various GBM models and neurodegeneration. Particular emphasis is also given to natural Hsp90 inhibitors that have been evidenced to show dual protection in both GBM and neurodegeneration.
Collapse
Affiliation(s)
- Sarmistha Mitra
- Department of Anatomy, Dongguk University College of Medicine, Gyeongju 38066, Republic of Korea
| | - Raju Dash
- Department of Anatomy, Dongguk University College of Medicine, Gyeongju 38066, Republic of Korea
| | - Yeasmin Akter Munni
- Department of Anatomy, Dongguk University College of Medicine, Gyeongju 38066, Republic of Korea
| | - Nusrat Jahan Selsi
- Product Development Department, Popular Pharmaceuticals Ltd., Dhaka 1207, Bangladesh
| | - Nasrin Akter
- Department of Clinical Pharmacy and Molecular Pharmacology, East West University Bangladesh, Dhaka 1212, Bangladesh
| | - Md Nazim Uddin
- Department of Pharmacy, Southern University Bangladesh, Chittagong 4000, Bangladesh
| | - Kishor Mazumder
- Department of Pharmacy, Jashore University of Science and Technology, Jashore 7408, Bangladesh
- School of Optometry and Vision Science, UNSW Medicine, University of New South Wales (UNSW), Sydney, NSW 2052, Australia
| | - Il Soo Moon
- Department of Anatomy, Dongguk University College of Medicine, Gyeongju 38066, Republic of Korea
| |
Collapse
|
11
|
Singh Tuli H, Rath P, Chauhan A, Sak K, Aggarwal D, Choudhary R, Sharma U, Vashishth K, Sharma S, Kumar M, Yadav V, Singh T, Yerer MB, Haque S. Luteolin, a Potent Anticancer Compound: From Chemistry to Cellular Interactions and Synergetic Perspectives. Cancers (Basel) 2022; 14:5373. [PMID: 36358791 PMCID: PMC9658186 DOI: 10.3390/cancers14215373] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 10/22/2022] [Accepted: 10/25/2022] [Indexed: 08/03/2023] Open
Abstract
Increasing rates of cancer incidence and the toxicity concerns of existing chemotherapeutic agents have intensified the research to explore more alternative routes to combat tumor. Luteolin, a flavone found in numerous fruits, vegetables, and herbs, has exhibited a number of biological activities, such as anticancer and anti-inflammatory. Luteolin inhibits tumor growth by targeting cellular processes such as apoptosis, cell-cycle progression, angiogenesis and migration. Mechanistically, luteolin causes cell death by downregulating Akt, PLK-1, cyclin-B1, cyclin-A, CDC-2, CDK-2, Bcl-2, and Bcl-xL, while upregulating BAX, caspase-3, and p21. It has also been reported to inhibit STAT3 signaling by the suppression of STAT3 activation and enhanced STAT3 protein degradation in various cancer cells. Therefore, extensive studies on the anticancer properties of luteolin reveal its promising role in chemoprevention. The present review describes all the possible cellular interactions of luteolin in cancer, along with its synergistic mode of action and nanodelivery insight.
Collapse
Affiliation(s)
- Hardeep Singh Tuli
- Department of Biotechnology, Maharishi Markandeshwar Engineering College, Maharishi Markandeshwar (Deemed to be University), Mullana-Ambala 133207, India
| | - Prangya Rath
- Amity Institute of Environmental Sciences, Amity University, Noida 201303, India
| | - Abhishek Chauhan
- Amity Institute of Environmental Toxicology, Safety and Management, Amity University, Noida 201303, India
| | | | - Diwakar Aggarwal
- Department of Biotechnology, Maharishi Markandeshwar Engineering College, Maharishi Markandeshwar (Deemed to be University), Mullana-Ambala 133207, India
| | - Renuka Choudhary
- Department of Biotechnology, Maharishi Markandeshwar Engineering College, Maharishi Markandeshwar (Deemed to be University), Mullana-Ambala 133207, India
| | - Ujjawal Sharma
- Department of Human Genetics and Molecular Medicine, Central University of Punjab, Bhatinda 151001, India
| | - Kanupriya Vashishth
- Department of Cardiology, Advance Cardiac Centre, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh 160012, India
| | - Sheetu Sharma
- Department of Pharmacovigilace and Clinical Research, Chitkara University, Rajpura 140401, India
| | - Manoj Kumar
- Department of Chemistry, Maharishi Markandeshwar University Sadopur, Ambala 133001, India
| | - Vikas Yadav
- Department of Translational Medicine, Clinical Research Centre, Skåne University Hospital, Lund University, SE-20213 Malmö, Sweden
| | - Tejveer Singh
- Translational Oncology Laboratory, Department of Zoology, Hansraj College, Delhi University, Delhi 110007, India
| | - Mukerrem Betul Yerer
- Department of Pharmacology, Faculty of Pharmacy, Erciyes University, Kayseri 38039, Turkey
| | - Shafiul Haque
- Research and Scientific Studies Unit, College of Nursing and Allied Health Sciences, Jazan University, Jazan 45142, Saudi Arabia
| |
Collapse
|
12
|
Diedrich C, Camargo Zittlau I, Schineider Machado C, Taise Fin M, Maissar Khalil N, Badea I, Mara Mainardes R. Mucoadhesive nanoemulsion enhances brain bioavailability of luteolin after intranasal administration and induces apoptosis to sh-sy5y neuroblastoma cells. Int J Pharm 2022; 626:122142. [PMID: 36064075 DOI: 10.1016/j.ijpharm.2022.122142] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 08/02/2022] [Accepted: 08/22/2022] [Indexed: 11/28/2022]
Abstract
Neuroblastoma is the most frequently diagnosed extracranial solid tumor in children and accounts for 7% of all childhood malignancies and 15% cancer mortality in children. Luteolin (LUT) is recognized by its anticancer activity against several types of cancer. The aim of this study was to prepare chitosan-coated nanoemulsion containing luteolin (NECh-LUT), investigate its potential for brain delivery following intranasal administration, and to evaluate its cytotoxicity against neuroblastoma cells. NECh-LUT was developed by cavitation process and characterized for its size, surface charge, encapsulation efficiency, and mucoadhesion. The developed formulation presented size 68±1 nm, zeta potential +13±1 mV, and encapsulation efficiency of 85.5±0.3%. The NECh-LUT presented nearly 6-fold higher permeation through the nasal mucosa ex vivo and prolonged LUT release up to 72 h in vitro, following Baker-Lonsdale kinetic model. The pharmacokinetic evaluation of NECh-LUT revealed a 10-fold increase in drug half-life and a 4.4 times enhancement in LUT biodistribution in brain tissue after intranasal administration of single-dose. In addition, NECh-LUT inhibited the growth of neuroblastoma cells after 24, 48 and 72 h in concentrations starting from 2 µM. The NECh-LUT developed for intranasal administration proved to be a promising alternative for brain delivery of LUT, and a viable option for the treatment of neuroblastoma.
Collapse
Affiliation(s)
- Camila Diedrich
- Pharmaceutical Nanotechnology Laboratory, Universidade Estadual do Centro-Oeste, Alameda Élio Antonio Dalla Vecchia, 838 - CEP 85040-167, Guarapuava, PR, Brazil
| | - Isabella Camargo Zittlau
- Pharmaceutical Nanotechnology Laboratory, Universidade Estadual do Centro-Oeste, Alameda Élio Antonio Dalla Vecchia, 838 - CEP 85040-167, Guarapuava, PR, Brazil
| | - Christiane Schineider Machado
- Pharmaceutical Nanotechnology Laboratory, Universidade Estadual do Centro-Oeste, Alameda Élio Antonio Dalla Vecchia, 838 - CEP 85040-167, Guarapuava, PR, Brazil
| | - Margani Taise Fin
- Pharmaceutical Nanotechnology Laboratory, Universidade Estadual do Centro-Oeste, Alameda Élio Antonio Dalla Vecchia, 838 - CEP 85040-167, Guarapuava, PR, Brazil
| | - Najeh Maissar Khalil
- Pharmaceutical Nanotechnology Laboratory, Universidade Estadual do Centro-Oeste, Alameda Élio Antonio Dalla Vecchia, 838 - CEP 85040-167, Guarapuava, PR, Brazil
| | - Ildiko Badea
- Drug Design and Discovery Research Group, College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, Canada
| | - Rubiana Mara Mainardes
- Pharmaceutical Nanotechnology Laboratory, Universidade Estadual do Centro-Oeste, Alameda Élio Antonio Dalla Vecchia, 838 - CEP 85040-167, Guarapuava, PR, Brazil.
| |
Collapse
|
13
|
Lee HS, Lee IH, Park SI, Jung M, Yang SG, Kwon TW, Lee DY. Unveiling the Mechanism of the Traditional Korean Medicinal Formula FDY003 on Glioblastoma Through a Computational Network Pharmacology Approach. Nat Prod Commun 2022. [DOI: 10.1177/1934578x221126311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Glioblastoma (GBM) is the most common type of primary malignant tumor that develops in the brain, with 0.21 million new cases per year globally and a median survival period of less than 2 years after diagnosis. Traditional Korean medicines have been increasingly suggested as effective and safe therapeutic strategies for GBM. However, their pharmacological effects and mechanistic characteristics remain to be studied. In this study, we employed a computational network pharmacological approach to determine the effects and mechanisms of the traditional Korean medicinal formula FDY003 on GBM. We found that FDY003 treatment decreased the viability of human GBM cells and increased their response to chemotherapeutics. We identified 10 potential active pharmacological compounds of FDY003 and 67 potential GBM-related target genes and proteins. The GBM-related targets of FDY003 were signaling components of various crucial GBM-associated pathways, such as PI3K-Akt, focal adhesion, MAPK, HIF-1, FoxO, Ras, and TNF. These pathways are functional regulators for the determination of cell growth and proliferation, survival and death, and cell division cycle of GBM cells. Together, the overall analyses contribute to the pharmacological basis for the anti-GBM roles of FDY003 and its systematic mechanisms.
Collapse
Affiliation(s)
- Ho-Sung Lee
- The Fore, Seoul, Republic of Korea
- Forest Hospital, Seoul, Republic of Korea
| | - In-Hee Lee
- The Fore, Seoul, Republic of Korea
- Forest Hospital, Seoul, Republic of Korea
| | | | - Minho Jung
- Forest Hospital, Seoul, Republic of Korea
| | | | | | - Dae-Yeon Lee
- The Fore, Seoul, Republic of Korea
- Forest Hospital, Seoul, Republic of Korea
| |
Collapse
|
14
|
Grabowska M, Kuczyński K, Piwecka M, Rabiasz A, Zemła J, Głodowicz P, Wawrzyniak D, Lekka M, Rolle K. miR-218 affects the ECM composition and cell biomechanical properties of glioblastoma cells. J Cell Mol Med 2022; 26:3913-3930. [PMID: 35702951 PMCID: PMC9279592 DOI: 10.1111/jcmm.17428] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 05/10/2022] [Accepted: 05/20/2022] [Indexed: 11/27/2022] Open
Abstract
Glioblastoma (GBM) is the most common malignant brain tumour. GBM cells have the ability to infiltrate into the surrounding brain tissue, which results in a significant decrease in the patient’s survival rate. Infiltration is a consequence of the low adhesion and high migration of the tumour cells, two features being associated with the highly remodelled extracellular matrix (ECM). In this study, we report that ECM composition is partially regulated at the post‐transcriptional level by miRNA. Particularly, we show that miR‐218, a well‐known miRNA suppressor, is involved in the direct regulation of ECM components, tenascin‐C (TN‐C) and syndecan‐2 (SDC‐2). We demonstrated that the overexpression of miR‐218 reduces the mRNA and protein expression levels of TN‐C and SDC‐2, and subsequently influences biomechanical properties of GBM cells. Atomic force microscopy (AFM) and real‐time migration analysis revealed that miR‐218 overexpression impairs the migration potential and enhances the adhesive properties of cells. AFM analysis followed by F‐actin staining demonstrated that the expression level of miR‐218 has an impact on cell stiffness and cytoskeletal reorganization. Global gene expression analysis showed deregulation of a number of genes involved in tumour cell motility and adhesion or ECM remodelling upon miR‐218 treatment, suggesting further indirect interactions between the cells and ECM. The results demonstrated a direct impact of miR‐218 reduction in GBM tumours on the qualitative ECM content, leading to changes in the rigidity of the ECM and GBM cells being conducive to increased invasiveness of GBM.
Collapse
Affiliation(s)
| | - Konrad Kuczyński
- Institute of Bioorganic Chemistry, Polish Academy of Sciences, Poznań, Poland.,NanoBioMedical Centre, Adam Mickiewicz University, Poznań, Poland
| | - Monika Piwecka
- Institute of Bioorganic Chemistry, Polish Academy of Sciences, Poznań, Poland
| | - Alicja Rabiasz
- Institute of Human Genetics, Polish Academy of Sciences, Poznań, Poland
| | - Joanna Zemła
- Institute of Nuclear Physics, Polish Academy of Sciences, Kraków, Poland
| | - Paweł Głodowicz
- Institute of Bioorganic Chemistry, Polish Academy of Sciences, Poznań, Poland
| | - Dariusz Wawrzyniak
- Institute of Bioorganic Chemistry, Polish Academy of Sciences, Poznań, Poland
| | - Małgorzata Lekka
- Institute of Nuclear Physics, Polish Academy of Sciences, Kraków, Poland
| | - Katarzyna Rolle
- Institute of Bioorganic Chemistry, Polish Academy of Sciences, Poznań, Poland
| |
Collapse
|
15
|
do Nascimento RP, dos Santos BL, Amparo JAO, Soares JRP, da Silva KC, Santana MR, Almeida ÁMAN, da Silva VDA, Costa MDFD, Ulrich H, Moura-Neto V, Lopes GPDF, Costa SL. Neuroimmunomodulatory Properties of Flavonoids and Derivates: A Potential Action as Adjuvants for the Treatment of Glioblastoma. Pharmaceutics 2022; 14:pharmaceutics14010116. [PMID: 35057010 PMCID: PMC8778519 DOI: 10.3390/pharmaceutics14010116] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 12/14/2021] [Accepted: 12/17/2021] [Indexed: 01/27/2023] Open
Abstract
Glioblastomas (GBMs) are tumors that have a high ability to migrate, invade and proliferate in the healthy tissue, what greatly impairs their treatment. These characteristics are associated with the complex microenvironment, formed by the perivascular niche, which is also composed of several stromal cells including astrocytes, microglia, fibroblasts, pericytes and endothelial cells, supporting tumor progression. Further microglia and macrophages associated with GBMs infiltrate the tumor. These innate immune cells are meant to participate in tumor surveillance and eradication, but they become compromised by GBM cells and exploited in the process. In this review we discuss the context of the GBM microenvironment together with the actions of flavonoids, which have attracted scientific attention due to their pharmacological properties as possible anti-tumor agents. Flavonoids act on a variety of signaling pathways, counteracting the invasion process. Luteolin and rutin inhibit NFκB activation, reducing IL-6 production. Fisetin promotes tumor apoptosis, while inhibiting ADAM expression, reducing invasion. Naringenin reduces tumor invasion by down-regulating metalloproteinases expression. Apigenin and rutin induce apoptosis in C6 cells increasing TNFα, while decreasing IL-10 production, denoting a shift from the immunosuppressive Th2 to the Th1 profile. Overall, flavonoids should be further exploited for glioma therapy.
Collapse
Affiliation(s)
- Ravena Pereira do Nascimento
- Laboratory of Neurochemistry and Cell Biology, Department of Biochemistry and Biophysics, Institute of Health Sciences, Federal University of Bahia, Salvador 40110-902, Bahia, Brazil; (R.P.d.N.); (B.L.d.S.); (J.A.O.A.); (J.R.P.S.); (K.C.d.S.); (M.R.S.); (Á.M.A.N.A.); (V.D.A.d.S.); (M.d.F.D.C.)
| | - Balbino Lino dos Santos
- Laboratory of Neurochemistry and Cell Biology, Department of Biochemistry and Biophysics, Institute of Health Sciences, Federal University of Bahia, Salvador 40110-902, Bahia, Brazil; (R.P.d.N.); (B.L.d.S.); (J.A.O.A.); (J.R.P.S.); (K.C.d.S.); (M.R.S.); (Á.M.A.N.A.); (V.D.A.d.S.); (M.d.F.D.C.)
- Academic College of Nurse, Department of Health, Federal University of Vale do São Francisco, Petrolina 56304-205, Pernambuco, Brazil
| | - Jéssika Alves Oliveira Amparo
- Laboratory of Neurochemistry and Cell Biology, Department of Biochemistry and Biophysics, Institute of Health Sciences, Federal University of Bahia, Salvador 40110-902, Bahia, Brazil; (R.P.d.N.); (B.L.d.S.); (J.A.O.A.); (J.R.P.S.); (K.C.d.S.); (M.R.S.); (Á.M.A.N.A.); (V.D.A.d.S.); (M.d.F.D.C.)
| | - Janaina Ribeiro Pereira Soares
- Laboratory of Neurochemistry and Cell Biology, Department of Biochemistry and Biophysics, Institute of Health Sciences, Federal University of Bahia, Salvador 40110-902, Bahia, Brazil; (R.P.d.N.); (B.L.d.S.); (J.A.O.A.); (J.R.P.S.); (K.C.d.S.); (M.R.S.); (Á.M.A.N.A.); (V.D.A.d.S.); (M.d.F.D.C.)
| | - Karina Costa da Silva
- Laboratory of Neurochemistry and Cell Biology, Department of Biochemistry and Biophysics, Institute of Health Sciences, Federal University of Bahia, Salvador 40110-902, Bahia, Brazil; (R.P.d.N.); (B.L.d.S.); (J.A.O.A.); (J.R.P.S.); (K.C.d.S.); (M.R.S.); (Á.M.A.N.A.); (V.D.A.d.S.); (M.d.F.D.C.)
| | - Monique Reis Santana
- Laboratory of Neurochemistry and Cell Biology, Department of Biochemistry and Biophysics, Institute of Health Sciences, Federal University of Bahia, Salvador 40110-902, Bahia, Brazil; (R.P.d.N.); (B.L.d.S.); (J.A.O.A.); (J.R.P.S.); (K.C.d.S.); (M.R.S.); (Á.M.A.N.A.); (V.D.A.d.S.); (M.d.F.D.C.)
| | - Áurea Maria Alves Nunes Almeida
- Laboratory of Neurochemistry and Cell Biology, Department of Biochemistry and Biophysics, Institute of Health Sciences, Federal University of Bahia, Salvador 40110-902, Bahia, Brazil; (R.P.d.N.); (B.L.d.S.); (J.A.O.A.); (J.R.P.S.); (K.C.d.S.); (M.R.S.); (Á.M.A.N.A.); (V.D.A.d.S.); (M.d.F.D.C.)
| | - Victor Diógenes Amaral da Silva
- Laboratory of Neurochemistry and Cell Biology, Department of Biochemistry and Biophysics, Institute of Health Sciences, Federal University of Bahia, Salvador 40110-902, Bahia, Brazil; (R.P.d.N.); (B.L.d.S.); (J.A.O.A.); (J.R.P.S.); (K.C.d.S.); (M.R.S.); (Á.M.A.N.A.); (V.D.A.d.S.); (M.d.F.D.C.)
| | - Maria de Fátima Dias Costa
- Laboratory of Neurochemistry and Cell Biology, Department of Biochemistry and Biophysics, Institute of Health Sciences, Federal University of Bahia, Salvador 40110-902, Bahia, Brazil; (R.P.d.N.); (B.L.d.S.); (J.A.O.A.); (J.R.P.S.); (K.C.d.S.); (M.R.S.); (Á.M.A.N.A.); (V.D.A.d.S.); (M.d.F.D.C.)
- National Institute for Translational Neurosciences (INCT/CNPq INNT), Rio de Janeiro 21941-902, Rio de Janeiro, Brazil;
| | - Henning Ulrich
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo 05508-000, São Paulo, Brazil
- Correspondence: (H.U.); (S.L.C.)
| | - Vivaldo Moura-Neto
- National Institute for Translational Neurosciences (INCT/CNPq INNT), Rio de Janeiro 21941-902, Rio de Janeiro, Brazil;
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo 05508-000, São Paulo, Brazil
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Rio de Janeiro, Brazil
- Paulo Niemeyer State Institute of the Brain, Rio de Janeiro 20230-024, Rio de Janeiro, Brazil
| | - Giselle Pinto de Faria Lopes
- Department of Marine Biotechnology, Admiral Paulo Moreira Institute for Sea Studies (IEAPM), Arraial do Cabo 28930-000, Rio de Janeiro, Brazil;
| | - Silvia Lima Costa
- Laboratory of Neurochemistry and Cell Biology, Department of Biochemistry and Biophysics, Institute of Health Sciences, Federal University of Bahia, Salvador 40110-902, Bahia, Brazil; (R.P.d.N.); (B.L.d.S.); (J.A.O.A.); (J.R.P.S.); (K.C.d.S.); (M.R.S.); (Á.M.A.N.A.); (V.D.A.d.S.); (M.d.F.D.C.)
- National Institute for Translational Neurosciences (INCT/CNPq INNT), Rio de Janeiro 21941-902, Rio de Janeiro, Brazil;
- Correspondence: (H.U.); (S.L.C.)
| |
Collapse
|
16
|
Alausa A, Victor UC, Celestine UO, Eweje IA, Balogun TA, Adeyemi R, Olatinwo M, Ogunlana AT, Oladipo O, Olaleke B. Phytochemical based sestrin2 pharmacological modulators in the treatment of adenocarcinomas. PHYTOMEDICINE PLUS 2021; 1:100133. [DOI: 10.1016/j.phyplu.2021.100133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
17
|
Hassan R, Mohi-Ud-Din R, Dar MO, Shah AJ, Mir PA, Shaikh M, Pottoo FH. Bioactive Heterocyclic Compounds as Potential Therapeutics in the Treatment of Gliomas: A Review. Anticancer Agents Med Chem 2021; 22:551-565. [PMID: 34488596 DOI: 10.2174/1871520621666210901112954] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 06/22/2021] [Accepted: 07/05/2021] [Indexed: 12/24/2022]
Abstract
Cancer is one of the most alarming diseases, with an estimation of 9.6 million deaths in 2018. Glioma occurs in glial cells surrounding nerve cells. The majority of the patients with gliomas have a terminal prognosis, and the ailment has significant sway on patients and their families, be it physical, psychological, or economic wellbeing. As Glioma exhibits, both intra and inter tumour heterogeneity with multidrug resistance and current therapies are ineffective. So the development of safer anti gliomas agents is the need of hour. Bioactive heterocyclic compounds, eithernatural or synthetic,are of potential interest since they have been active against different targets with a wide range of biological activities, including anticancer activities. In addition, they can cross the biological barriers and thus interfere with various signalling pathways to induce cancer cell death. All these advantages make bioactive natural compounds prospective candidates in the management of glioma. In this review, we assessed various bioactive heterocyclic compounds, such as jaceosidin, hispudlin, luteolin, silibinin, cannabidiol, tetrahydrocannabinol, didemnin B, thymoquinone, paclitaxel, doxorubicin, and cucurbitacins for their potential anti-glioma activity. Also, different kinds of chemical reactions to obtain various heterocyclic derivatives, e.g. indole, indazole, benzimidazole, benzoquinone, quinoline, quinazoline, pyrimidine, and triazine, are listed.
Collapse
Affiliation(s)
- Reyaz Hassan
- Pharmaceutical Chemistry Division, Department of Pharmaceutical Sciences, University of Kashmir, Hazratbal, Srinagar-190006, Kashmir. India
| | - Roohi Mohi-Ud-Din
- Pharmacognosy Division, Department of Pharmaceutical Sciences, University of Kashmir, Hazratbal, Srinagar, 190006, Kashmir. India
| | - Mohammad Ovais Dar
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Science and Research (NIPER), S.A.S. Nagar, Mohali, Punjab-160062. India
| | - Abdul Jalil Shah
- Pharmaceutical Chemistry Division, Department of Pharmaceutical Sciences, University of Kashmir, Hazratbal, Srinagar-190006, Kashmir. India
| | - Prince Ahad Mir
- Amritsar Pharmacy College, 12 KM stone Amritsar Jalandhar GT Road, Mandwala-143001. India
| | - Majeed Shaikh
- Natural Product Chemistry Division, CSIR-Indian Institute of Integrative Medicine, Canal Road, Jammu-180001. India
| | - Faheem Hyder Pottoo
- Department of Pharmacology, College of Clinical Pharmacy, Imam Abdulrahman Bin Faisal University, 31441, Dammam. Saudi Arabia
| |
Collapse
|
18
|
Cotoraci C, Ciceu A, Sasu A, Miutescu E, Hermenean A. Bioactive Compounds from Herbal Medicine Targeting Multiple Myeloma. APPLIED SCIENCES 2021; 11:4451. [DOI: 10.3390/app11104451] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Abstract
Multiple myeloma (MM) is one of the most widespread hematological cancers. It is characterized by a clonal proliferation of malignant plasma cells in the bone marrow and by the overproduction of monoclonal proteins. In recent years, the survival rate of patients with multiple myeloma has increased significantly due to the use of transplanted stem cells and of the new therapeutic agents that have significantly increased the survival rate, but it still cannot be completely cured and therefore the development of new therapeutic products is needed. Moreover, many patients have various side effects and face the development of drug resistance to current therapies. The purpose of this review is to highlight the bioactive active compounds (flavonoids) and herbal extracts which target dysregulated signaling pathway in MM, assessed by in vitro and in vivo experiments or clinical studies, in order to explore their healing potential targeting multiple myeloma. Mechanistically, they demonstrated the ability to promote cell cycle blockage and apoptosis or autophagy in cancer cells, as well as inhibition of proliferation/migration/tumor progression, inhibition of angiogenesis in the tumor vascular network. Current research provides valuable new information about the ability of flavonoids to enhance the apoptotic effects of antineoplastic drugs, thus providing viable therapeutic options based on combining conventional and non-conventional therapies in MM therapeutic protocols.
Collapse
Affiliation(s)
- Coralia Cotoraci
- Department of Hematology, Faculty of Medicine, Vasile Goldis Western University of Arad, Rebreanu 86, 310414 Arad, Romania
| | - Alina Ciceu
- “Aurel Ardelean” Institute of Life Sciences, Vasile Godis Western University of Arad, Rebreanu 86, 310414 Arad, Romania
| | - Alciona Sasu
- Department of Hematology, Faculty of Medicine, Vasile Goldis Western University of Arad, Rebreanu 86, 310414 Arad, Romania
| | - Eftimie Miutescu
- Department of Gastroenterology, Faculty of Medicine, Vasile Goldis Western University of Arad, Rebreanu 86, 310414 Arad, Romania
| | - Anca Hermenean
- “Aurel Ardelean” Institute of Life Sciences, Vasile Godis Western University of Arad, Rebreanu 86, 310414 Arad, Romania
- Department of Histology, Faculty of Medicine, Vasile Goldis Western University of Arad, Rebreanu 86, 310414 Arad, Romania
| |
Collapse
|
19
|
Glogowska A, Thanasupawat T, Beiko J, Pitz M, Hombach-Klonisch S, Klonisch T. Novel CTRP8-RXFP1-JAK3-STAT3 axis promotes Cdc42-dependent actin remodeling for enhanced filopodia formation and motility in human glioblastoma cells. Mol Oncol 2021; 16:368-387. [PMID: 33960104 PMCID: PMC8763656 DOI: 10.1002/1878-0261.12981] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Revised: 03/23/2021] [Accepted: 05/04/2021] [Indexed: 12/14/2022] Open
Abstract
C1q tumor necrosis factor‐related peptide 8 (CTRP8) is the least studied member of the C1Q‐TNF‐related peptide family. We identified CTRP8 as a ligand of the G protein‐coupled receptor relaxin family peptide receptor 1 (RXFP1) in glioblastoma multiforme (GBM). The CTRP8‐RXFP1 ligand–receptor system protects human GBM cells against the DNA‐alkylating damage‐inducing temozolomide (TMZ), the drug of choice for the treatment of patients with GBM. The DNA protective role of CTRP8 was dependent on a functional RXFP1‐STAT3 signaling cascade and targeted the monofunctional glycosylase N‐methylpurine DNA glycosylase (MPG) for more efficient base excision repair of TMZ‐induced DNA‐damaged sites. CTRP8 also improved the survival of GBM cells by upregulating anti‐apoptotic BCl‐2 and BCL‐XL. Here, we have identified Janus‐activated kinase 3 (JAK3) as a novel member of a novel CTRP8‐RXFP1‐JAK3‐STAT3 signaling cascade that caused an increase in cellular protein content and activity of the small Rho GTPase Cdc42. This is associated with significant F‐actin remodeling and increased GBM motility. Cdc42 was critically important for the upregulation of the actin nucleation complex N‐Wiskott–Aldrich syndrome protein/Arp3/4 and actin elongation factor profilin‐1. The activation of the RXFP1‐JAK3‐STAT3‐Cdc42 axis by both RXFP1 agonists, CTRP8 and relaxin‐2, caused extensive filopodia formation. This coincided with enhanced activity of ezrin, a key factor in tethering F‐actin to the plasma membrane, and inhibition of the actin filament severing activity of cofilin. The F‐actin remodeling and pro‐migratory activities promoted by the novel RXFP1‐JAK3‐STAT3‐Cdc42 axis were blocked by JAK3 inhibitor tofacitinib and STAT3 inhibitor STAT3 inhibitor VI. This provides a new rationale for the design of JAK3 and STAT3 inhibitors with better brain permeability for clinical treatment of the pervasive brain invasiveness of GBM.
Collapse
Affiliation(s)
- Aleksandra Glogowska
- Department of Human Anatomy and Cell Science, Rady Faculty of Health Sciences, College of Medicine, University of Manitoba, Winnipeg, Canada
| | - Thatchawan Thanasupawat
- Department of Human Anatomy and Cell Science, Rady Faculty of Health Sciences, College of Medicine, University of Manitoba, Winnipeg, Canada
| | - Jason Beiko
- Department of Surgery, Rady Faculty of Health Sciences, College of Medicine, University of Manitoba, Winnipeg, Canada
| | - Marshall Pitz
- Research Institute in Oncology and Hematology (RIOH), CancerCare Manitoba, Rady Faculty of Health Sciences, College of Medicine, University of Manitoba, Winnipeg, Canada
| | - Sabine Hombach-Klonisch
- Department of Human Anatomy and Cell Science, Rady Faculty of Health Sciences, College of Medicine, University of Manitoba, Winnipeg, Canada.,Research Institute in Oncology and Hematology (RIOH), CancerCare Manitoba, Rady Faculty of Health Sciences, College of Medicine, University of Manitoba, Winnipeg, Canada
| | - Thomas Klonisch
- Department of Human Anatomy and Cell Science, Rady Faculty of Health Sciences, College of Medicine, University of Manitoba, Winnipeg, Canada.,Department of Surgery, Rady Faculty of Health Sciences, College of Medicine, University of Manitoba, Winnipeg, Canada.,Research Institute in Oncology and Hematology (RIOH), CancerCare Manitoba, Rady Faculty of Health Sciences, College of Medicine, University of Manitoba, Winnipeg, Canada.,Department of Medical Microbiology & Infectious Diseases, Rady Faculty of Health Sciences, College of Medicine, University of Manitoba, Winnipeg, Canada.,Department of Pathology, Rady Faculty of Health Sciences, College of Medicine, University of Manitoba, Winnipeg, Canada
| |
Collapse
|
20
|
Moghadam FA, Dabirian S, Dogaheh MG, Mojabi M, Yousefbeyk F, Ghasemi S. Novel 4-Anilinoquinazoline Derivatives as Potent Anticancer Agents: Design, Synthesis, Cytotoxic Activity, and Docking Study. Aust J Chem 2021. [DOI: 10.1071/ch21147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
The simultaneous inhibition of EGFR and VEGFR-2 is a promising method in cancer treatment. In the present work, several 4-anilinoquinazoline derivatives encompassing different substitutions at the C-4 and C-7 positions of a quinazoline core were designed, synthesised, and evaluated for their cytotoxicity on A431, HUVEC, and HU02 cell lines. Docking studies were carried out to test the interactions of all synthesised compounds with EGFR and VEGFR-2. Furthermore, a wound healing assay was done for the investigation of cell migration. The most potent compound was 8l followed by the compounds 8i and 8j which showed better cytotoxic activities on A431 and HUVEC cell lines than the standard (Vandetanib). The compounds 8f and 8a represented the best docking energies of 8.99 and 9.35 kcal mol−1 for EGFR and VEGFR, respectively. Moreover, molecular docking studies exhibited that compound 8l showed efficient binding affinity against both EGFR and VEGFR-2. It can bind to these receptors through the formation of essential hydrogen bonds between the quinazoline N1 atom and the Met796 backbone of EGFR and two hydrogen bonds with Cys919 and Thr916 of VEGFR-2 with energies of –7.99 and –7.85 kcal mol−1, respectively. In addition, this compound displayed the highest activity on cell migration and wound healing. Compound 8l with the highest cytotoxic activity can be considered a candidate for further investigation and structural optimisation as an antiproliferative agent.
Collapse
|
21
|
Reddy MN, Adnan M, Alreshidi MM, Saeed M, Patel M. Evaluation of Anticancer, Antibacterial and Antioxidant Properties of a Medicinally Treasured Fern Tectaria coadunata with its Phytoconstituents Analysis by HR-LCMS. Anticancer Agents Med Chem 2020; 20:1845-1856. [DOI: 10.2174/1871520620666200318101938] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 02/11/2020] [Accepted: 02/18/2020] [Indexed: 02/06/2023]
Abstract
Background:
Tectaria coadunata (T. coadunata) is an important fern species with a number of
medicinal properties. It has been evidently found for its effectiveness in ethanomedicinal usage, which can also
emerge as one of the most promising sources for nutraceuticals.
Objective:
This study aims to examine the phytochemistry of the whole crude extract of T. coadunata for the
first time with evaluation of antibacterial, antioxidant and anticancer activity.
Methods:
High Resolution Liquid Chromatography Mass Spectrometry analysis (HR-LCMS) was performed for
confirming the presence of biologically active constituents in the extract of T. coadunata followed by antibacterial,
antioxidant and anticancer activity.
Results:
With the detailed Mass spectra data, absorbance spectra and retention times, chemical composition of
T. coadunata holds a diverse group of bioactive/chemical components such as sugars, sugar alcohol, flavonoids,
terpenoids and phenolics. The results for antioxidant activity showed that T. coadunata crude extract had higher
scavenging potential against 2,2-diphenyl-1-picrylhydrazyl (DPPH) free radicals than H2O2 molecules, which
was followed by positive antibacterial activity against several pathogenic bacteria like Shigella flexneri, Staphylococcus
aureus and Salmonella typhi.
Discussion:
The ethanolic extract of T. coadunata showed favorable antiproliferation activity against three
leukemic (KG1, MOLT-3 and K-562) cells in a dose dependent manner, especially for KG1 42.850±1.24μg/ml.
Conclusion:
This study has provided a better understanding of the presence of biologically active phytochemical
constituents in the extract of T. coadunata, which can be the reason for its bioactive potential. Moreover,
T. coadunata has significant anticancer activities against human leukemic cancer cell lines, indicating it as a potential
anticancer agent.
Collapse
Affiliation(s)
- Mandadi N. Reddy
- Bapalal Vaidya Botanical Research Centre, Department of Biosciences, Veer Narmad South Gujarat University, Surat, Gujarat, India
| | - Mohd. Adnan
- Department of Biology, College of Science, University of Ha’il, Ha’il, PO Box 2440, Saudi Arabia
| | - Mousa M. Alreshidi
- Department of Biology, College of Science, University of Ha’il, Ha’il, PO Box 2440, Saudi Arabia
| | - Mohd. Saeed
- Department of Biology, College of Science, University of Ha’il, Ha’il, PO Box 2440, Saudi Arabia
| | - Mitesh Patel
- Bapalal Vaidya Botanical Research Centre, Department of Biosciences, Veer Narmad South Gujarat University, Surat, Gujarat, India
| |
Collapse
|
22
|
Xiang C, Wu X, Zhao Z, Feng X, Bai X, Liu X, Zhao J, Takeda S, Qing Y. Nonhomologous end joining and homologous recombination involved in luteolin-induced DNA damage in DT40 cells. Toxicol In Vitro 2020; 65:104825. [PMID: 32169435 DOI: 10.1016/j.tiv.2020.104825] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 02/08/2020] [Accepted: 03/08/2020] [Indexed: 02/05/2023]
Abstract
Luteolin (3',4',5,7-tetrahydroxyflavone), a naturally occurring flavonoid, has been shown to have anticancer activity in many types of cancer cell lines. The anticancer capacity of luteolin may be related to its ability to induce DNA double-strand breaks (DSBs). Here, we used DT40 cells to determine whether nonhomologous end joining (NHEJ) and homologous recombination (HR) are involved in the repair mechanism of luteolin-induced DNA damage. Cells defective in Ku70 (an enzyme associated with NHEJ) or Rad54 (an enzyme essential for HR) were hypersensitive and presented more apoptosis in response to luteolin. Moreover, the sensitivity and apoptosis of Ku70-/- and Rad54-/- cells were associated with increased DNA damage when the numbers of γ-H2AX foci and chromosomal aberrations (CAs) were compared with those from WT cells. Additionally, after treatment with luteolin, Ku70-/- cells presented more Top2 covalent cleavage complexes (Top2cc). These results indicated that luteolin induced DSBs in DT40 cells and demonstrated that both NHEJ and HR participated in the repair of luteolin-induced DSBs, which might be related to the inhibition of topoisomerases. These results imply that simultaneous inhibition of NHEJ and HR with luteolin treatment would provide a powerful protocol in cancer chemotherapy.
Collapse
Affiliation(s)
- Cuifang Xiang
- Department of Pharmacology, Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, West China School of Pharmacy, Sichuan University, Chengdu, Sichuan, China
| | - Xiaohua Wu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Zilu Zhao
- Department of Pharmacology, Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, West China School of Pharmacy, Sichuan University, Chengdu, Sichuan, China
| | - Xiaoyu Feng
- Department of Pharmacology, Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, West China School of Pharmacy, Sichuan University, Chengdu, Sichuan, China
| | - Xin Bai
- Department of Pharmacology, Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, West China School of Pharmacy, Sichuan University, Chengdu, Sichuan, China
| | - Xin Liu
- Department of Pharmacology, Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, West China School of Pharmacy, Sichuan University, Chengdu, Sichuan, China
| | - Jingxia Zhao
- Department of Pharmacology, Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, West China School of Pharmacy, Sichuan University, Chengdu, Sichuan, China
| | - Shunichi Takeda
- Department of Radiation Genetics, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Yong Qing
- Department of Pharmacology, Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, West China School of Pharmacy, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
23
|
Mirza-Aghazadeh-Attari M, Ekrami EM, Aghdas SAM, Mihanfar A, Hallaj S, Yousefi B, Safa A, Majidinia M. Targeting PI3K/Akt/mTOR signaling pathway by polyphenols: Implication for cancer therapy. Life Sci 2020; 255:117481. [PMID: 32135183 DOI: 10.1016/j.lfs.2020.117481] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2019] [Revised: 02/20/2020] [Accepted: 02/28/2020] [Indexed: 02/07/2023]
Abstract
Cancer is one of the biggest challenges facing medicine and its cure is regarded to be the Holy Grail of medicine. Therapy in cancer is consisted as various artificial cytotoxic agents and radiotherapy, and recently immunotherapy. Recently much attention has been directed to the use of natural occurring agents in cancer therapy. One of the main group of agents utilized in this regard is polyphenols which are found abundantly in berries, fruits and vegetables. Polyphenols show to exert direct and indirect effects in progression of cancer, angiogenesis, proliferation and enhancing resistance to treatment. One of the cellular pathways commonly affected by polyphenols is PI3K/Akt/mTOR pathway, which has far ranging effects on multiple key aspects of cellular growth, metabolism and death. In this review article, evidence regarding the biology of polyphenols in cancer via PI3K/Akt/mTOR pathway is discussed and their application on cancer pathophysiology in various types of human malignancies is shown.
Collapse
Affiliation(s)
- Mohammad Mirza-Aghazadeh-Attari
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran; Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Elyad Mohammadi Ekrami
- Department of Anesthesiology & Critical Care Medicine, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Seyyed Ali Mousavi Aghdas
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran; Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ainaz Mihanfar
- Department of Biochemistry, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Shahin Hallaj
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran; Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Bahman Yousefi
- Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amin Safa
- Institute of Research and Development, Duy Tan University, Da Nang, Viet Nam; Department of Immunology, Ophthalmology and ENT, School of Medicine, Complutense University, Madrid, Spain
| | - Maryam Majidinia
- Solid Tumor Research Center, Urmia University of Medical Sciences, Urmia, Iran.
| |
Collapse
|
24
|
The Role of Rho GTPases in Motility and Invasion of Glioblastoma Cells. Anal Cell Pathol (Amst) 2020; 2020:9274016. [PMID: 32089990 PMCID: PMC7013281 DOI: 10.1155/2020/9274016] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2019] [Revised: 01/14/2020] [Accepted: 01/22/2020] [Indexed: 12/27/2022] Open
Abstract
Astrocytomas are primary malignant brain tumors that originate from astrocytes. Grade IV astrocytoma or glioblastoma is a highly invasive tumor that occur within the brain parenchyma. The Rho family of small GTPases, which includes Rac1, Cdc42, and RhoA, is an important family whose members are key regulators of the invasion and migration of glioblastoma cells. In this review, we describe the role played by the Rho family of GTPases in the regulation of the invasion and migration of glioblastoma cells. Specifically, we focus on the role played by RhoA, Rac1, RhoG, and Cdc42 in cell migration through rearrangement of actin cytoskeleton, cell adhesion, and invasion. Finally, we highlight the importance of potentially targeting Rho GTPases in the treatment of glioblastoma.
Collapse
|
25
|
Imran M, Rauf A, Abu-Izneid T, Nadeem M, Shariati MA, Khan IA, Imran A, Orhan IE, Rizwan M, Atif M, Gondal TA, Mubarak MS. Luteolin, a flavonoid, as an anticancer agent: A review. Biomed Pharmacother 2019; 112:108612. [PMID: 30798142 DOI: 10.1016/j.biopha.2019.108612] [Citation(s) in RCA: 527] [Impact Index Per Article: 87.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 01/06/2019] [Accepted: 01/23/2019] [Indexed: 10/27/2022] Open
Abstract
Many food-derived phytochemicals and their derivatives represent a cornucopia of new anti-cancer compounds. Luteolin (3,4,5,7-tetrahydroxy flavone) is a flavonoid found in different plants such as vegetables, medicinal herbs, and fruits. It acts as an anticancer agent against various types of human malignancies such as lung, breast, glioblastoma, prostate, colon, and pancreatic cancers. It also blocks cancer development in vitro and in vivo by inhibition of proliferation of tumor cells, protection from carcinogenic stimuli, and activation of cell cycle arrest, and by inducing apoptosis through different signaling pathways. Luteolin can additionally reverse epithelial-mesenchymal transition (EMT) through a mechanism that involves cytoskeleton shrinkage, induction of the epithelial biomarker E-cadherin expression, and by down-regulation of the mesenchymal biomarkers N-cadherin, snail, and vimentin. Furthermore, luteolin increases levels of intracellular reactive oxygen species (ROS) by activation of lethal endoplasmic reticulum stress response and mitochondrial dysfunction in glioblastoma cells, and by activation of ER stress-associated proteins expressions, including phosphorylation of eIF2α, PERK, CHOP, ATF4, and cleaved-caspase 12. Accordingly, the present review article summarizes the progress of recent research on luteolin against several human cancers.
Collapse
Affiliation(s)
- Muhammad Imran
- University Institute of Diet and Nutritional Sciences, Faculty of Allied Health Sciences, The University of Lahor, Pakistan
| | - Abdur Rauf
- Department of Chemistry, University of Swabi, Anbar, 23561, Khyber Pakhtunkhwa, Pakistan.
| | - Tareq Abu-Izneid
- Department of Pharmaceutical Sciences, College of Pharmacy, Al Ain University of Science and Technology, Al Ain Campus, UAE
| | - Muhammad Nadeem
- Department of Environmental Sciences, COMSATS Institute of Information Technology, Vehari, Pakistan
| | - Mohammad Ali Shariati
- Laboratory of Biocontrol and Antimicrobial Resistance, Orel State, University Named After I.S. Turgenev, 302026, Orel, Russia
| | - Imtiaz Ali Khan
- Department of Agriculture, University of Swabi, Anbar, 23561, Khyber Pakhtunkhwa, Pakistan
| | - Ali Imran
- Department of Food Science, Nutrition & Home Economics, Institute of Home and Food Sciences, Government College University, Faisalabad, Pakistan
| | - Ilkay Erdogan Orhan
- Department of Pharmacognosy, Faculty of Pharmacy, Gazi University, 06330, Ankara, Turkey.
| | - Muhammad Rizwan
- Department of Microbiology and Biotechnology, Abasyn University Peshawar, KPK, Pakistan
| | - Muhammad Atif
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Sakaka, Saudi Arabia
| | - Tanweer Aslam Gondal
- School of Exercise and Nutrition, Faculty of Health, Deakin University, Australia
| | - Mohammad S Mubarak
- Department of Chemistry, The University of Jordan, Amman, 11942, Jordan.
| |
Collapse
|
26
|
Yi C, Li G, Ivanov DN, Wang Z, Velasco MX, Hernández G, Kaundal S, Villarreal J, Gupta YK, Qiao M, Hubert CG, Hart MJ, Penalva LOF. Luteolin inhibits Musashi1 binding to RNA and disrupts cancer phenotypes in glioblastoma cells. RNA Biol 2018; 15:1420-1432. [PMID: 30362859 DOI: 10.1080/15476286.2018.1539607] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
RNA binding proteins have emerged as critical oncogenic factors and potential targets in cancer therapy. In this study, we evaluated Musashi1 (Msi1) targeting as a strategy to treat glioblastoma (GBM); the most aggressive brain tumor type. Msi1 expression levels are often high in GBMs and other tumor types and correlate with poor clinical outcome. Moreover, Msi1 has been implicated in chemo- and radio-resistance. Msi1 modulates a range of cancer relevant processes and pathways and regulates the expression of stem cell markers and oncogenic factors via mRNA translation/stability. To identify Msi1 inhibitors capable of blocking its RNA binding function, we performed a ~ 25,000 compound fluorescence polarization screen. NMR and LSPR were used to confirm direct interaction between Msi1 and luteolin, the leading compound. Luteolin displayed strong interaction with Msi1 RNA binding domain 1 (RBD1). As a likely consequence of this interaction, we observed via western and luciferase assays that luteolin treatment diminished Msi1 positive impact on the expression of pro-oncogenic target genes. We tested the effect of luteolin treatment on GBM cells and showed that it reduced proliferation, cell viability, colony formation, migration and invasion of U251 and U343 GBM cells. Luteolin also decreased the proliferation of patient-derived glioma initiating cells (GICs) and tumor-organoids but did not affect normal astrocytes. Finally, we demonstrated the value of combined treatments with luteolin and olaparib (PARP inhibitor) or ionizing radiation (IR). Our results show that luteolin functions as an inhibitor of Msi1 and demonstrates its potential use in GBM therapy.
Collapse
Affiliation(s)
- Caihong Yi
- a Greehey Children's Cancer Research Institute , University of Texas Health Science Center , San Antonio , TX , USA.,b Xiangya School of Medicine , Central South University , Hunan , China
| | - Guiming Li
- c Center for Innovative Drug Discovery , University of Texas Health Science Center , San Antonio , TX , USA.,d Department of Biochemistry and Structural Biology , University of Texas Health Science Center , San Antonio , TX , USA
| | - Dmitri N Ivanov
- d Department of Biochemistry and Structural Biology , University of Texas Health Science Center , San Antonio , TX , USA
| | - Zhonghua Wang
- d Department of Biochemistry and Structural Biology , University of Texas Health Science Center , San Antonio , TX , USA
| | - Mitzli X Velasco
- a Greehey Children's Cancer Research Institute , University of Texas Health Science Center , San Antonio , TX , USA.,e Division of Basic Research , National Institute of Cancer (INCan) , Mexico City , Mexico
| | - Greco Hernández
- e Division of Basic Research , National Institute of Cancer (INCan) , Mexico City , Mexico
| | - Soni Kaundal
- a Greehey Children's Cancer Research Institute , University of Texas Health Science Center , San Antonio , TX , USA
| | - Johanna Villarreal
- a Greehey Children's Cancer Research Institute , University of Texas Health Science Center , San Antonio , TX , USA
| | - Yogesh K Gupta
- a Greehey Children's Cancer Research Institute , University of Texas Health Science Center , San Antonio , TX , USA.,d Department of Biochemistry and Structural Biology , University of Texas Health Science Center , San Antonio , TX , USA
| | - Mei Qiao
- a Greehey Children's Cancer Research Institute , University of Texas Health Science Center , San Antonio , TX , USA
| | - Christopher G Hubert
- f Department of Stem Cell Biology and Regenerative Medicine , Cleveland Clinic , Cleveland , OH , USA
| | - Matthew J Hart
- a Greehey Children's Cancer Research Institute , University of Texas Health Science Center , San Antonio , TX , USA.,c Center for Innovative Drug Discovery , University of Texas Health Science Center , San Antonio , TX , USA.,d Department of Biochemistry and Structural Biology , University of Texas Health Science Center , San Antonio , TX , USA
| | - Luiz O F Penalva
- a Greehey Children's Cancer Research Institute , University of Texas Health Science Center , San Antonio , TX , USA.,g Department of Cell Systems and Anatomy , University of Texas Health Science Center , San Antonio , TX , USA
| |
Collapse
|
27
|
Erices JI, Torres Á, Niechi I, Bernales I, Quezada C. Current natural therapies in the treatment against glioblastoma. Phytother Res 2018; 32:2191-2201. [PMID: 30109743 DOI: 10.1002/ptr.6170] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2018] [Revised: 06/08/2018] [Accepted: 07/03/2018] [Indexed: 12/11/2022]
Abstract
Glioblastoma (GBM) is the most common and aggressive brain tumor, which causes the highest number of deaths worldwide. It is a highly vascularized tumor, infiltrative, and its tumorigenic capacity is exacerbated. All these hallmarks are therapeutic targets in GBM treatment, including surgical removal followed by radiotherapy and chemotherapy. Current therapies have not been sufficient for the effective patient's management, so the classic therapies have had to expand and incorporate new alternative treatments, including natural compounds. This review summarizes natural products and their physiological effects in in vitro and in vivo models of GBM, specifically by modulating signaling pathways involved in angiogenesis, cell migration/invasion, cell viability, apoptosis, and chemoresistance. The most important aspects of natural products and their derivatives were described in relation to its antitumoral effects. As a final result, it can be obtained that within the compounds with more evidence that supports or suggests its clinical use are the cannabinoids, terpenes, and curcumin, because many have been shown to have a significant effect in decreasing the progress of GBM through known mechanisms, such as chemo-sensitization or decrease migration and cell invasion. Natural compounds emerge as promising therapies to attack the progress of GBM.
Collapse
Affiliation(s)
- José Ignacio Erices
- Instituto de Bioquímica y Microbiología, Facultad de Ciencias, Universidad Austral de Chile, Valdivia, Chile
| | - Ángelo Torres
- Instituto de Bioquímica y Microbiología, Facultad de Ciencias, Universidad Austral de Chile, Valdivia, Chile
| | - Ignacio Niechi
- Instituto de Bioquímica y Microbiología, Facultad de Ciencias, Universidad Austral de Chile, Valdivia, Chile
| | - Isabel Bernales
- Instituto de Bioquímica y Microbiología, Facultad de Ciencias, Universidad Austral de Chile, Valdivia, Chile
| | - Claudia Quezada
- Instituto de Bioquímica y Microbiología, Facultad de Ciencias, Universidad Austral de Chile, Campus Isla Teja s/n, Valdivia, Chile
| |
Collapse
|
28
|
Pu Y, Zhang T, Wang J, Mao Z, Duan B, Long Y, Xue F, Liu D, Liu S, Gao Z. Luteolin exerts an anticancer effect on gastric cancer cells through multiple signaling pathways and regulating miRNAs. J Cancer 2018; 9:3669-3675. [PMID: 30405835 PMCID: PMC6215998 DOI: 10.7150/jca.27183] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Accepted: 08/02/2018] [Indexed: 12/16/2022] Open
Abstract
Accumulating studies confirmed that luteolin, a common dietary flavonoid which is widely distributed in plants and has diverse beneficial biological function, including anti-oxidant, anti-inflammation and anticancer properties. However, the detail mechanisms of luteolin on GC are poorly understood. Here, we investigated the anticancer effect of luteolin in GC cells in vitro and in vivo. Luteolin reduced the cell viability in a time and dose-dependent manner. Luteolin significantly inhibited cell cycle progress, colony formation, proliferation, migration, invasion and promoted apoptosis in vitro and in vivo. Luteolin also regulated these biological effects associated regulators. Mechanically, luteolin treatment regulated Notch1, PI3K, AKT, mTOR, ERK, STAT3 and P38 signaling pathways and modulated a series of miRNAs expression. These findings provide novel insight into the molecular function of luteolin which suggest its potential as a therapeutic agent for human GC.
Collapse
Affiliation(s)
- Yansong Pu
- The Second Department of General Surgery, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi 710068, P.R. China
| | - Tao Zhang
- The Second Department of General Surgery, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi 710068, P.R. China
| | - Jianhua Wang
- The Second Department of General Surgery, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi 710068, P.R. China
| | - Zhijun Mao
- The Second Department of General Surgery, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi 710068, P.R. China
| | - Baojun Duan
- Department of Oncology, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi 710068, P.R. China
| | - Yanbin Long
- The Second Department of General Surgery, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi 710068, P.R. China
| | - Fei Xue
- The Second Department of General Surgery, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi 710068, P.R. China
| | - Dong Liu
- The Second Department of General Surgery, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi 710068, P.R. China
| | - Sida Liu
- The Second Department of General Surgery, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi 710068, P.R. China
| | - Zengzhan Gao
- The Second Department of General Surgery, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi 710068, P.R. China
| |
Collapse
|
29
|
Anson DM, Wilcox RM, Huseman ED, Stump TA, Paris RL, Darkwah BO, Lin S, Adegoke AO, Gryka RJ, Jean-Louis DS, Amos S. Luteolin Decreases Epidermal Growth Factor Receptor-Mediated Cell Proliferation and Induces Apoptosis in Glioblastoma Cell Lines. Basic Clin Pharmacol Toxicol 2018; 123:678-686. [PMID: 29935053 DOI: 10.1111/bcpt.13077] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Accepted: 06/20/2018] [Indexed: 12/11/2022]
Abstract
Glioblastomas are a subtype of gliomas, which are the most aggressive and deadly form of brain tumours. The epidermal growth factor receptor (EGFR) is over-expressed and amplified in glioblastomas. Luteolin is a common bioflavonoid found in a variety of fruits and vegetables. The aim of this study was to explore the molecular and biological effects of luteolin on EGF-induced cell proliferation and the potential of luteolin to induce apoptosis in glioblastoma cells. In vitro cell viability assays demonstrated that luteolin decreased cell proliferation in the presence or absence of EGF. Immunoblots revealed that luteolin decreased the protein expression levels of phosphorylated Akt, mTOR, p70S6K and MAPK in the presence of EGF. Furthermore, our results revealed the ability of luteolin to induce caspase and PARP cleavages in glioblastoma cells in addition to promoting cell cycle arrest. Our results demonstrated that luteolin has an inhibitory effect on downstream signalling molecules activated by EGFR, particularly the Akt and MAPK signalling pathways, and provided a rationale for further clinical investigation into the use of luteolin as a therapeutic molecule in the management of glioblastoma.
Collapse
Affiliation(s)
- David M Anson
- Department of Pharmaceutical Sciences, School of Pharmacy, Cedarville University, Cedarville, OH, USA
| | - Rachel M Wilcox
- Department of Pharmaceutical Sciences, School of Pharmacy, Cedarville University, Cedarville, OH, USA
| | - Eric D Huseman
- Department of Pharmaceutical Sciences, School of Pharmacy, Cedarville University, Cedarville, OH, USA
| | - Trevor A Stump
- Department of Pharmaceutical Sciences, School of Pharmacy, Cedarville University, Cedarville, OH, USA
| | - Robert L Paris
- Department of Pharmaceutical Sciences, School of Pharmacy, Cedarville University, Cedarville, OH, USA
| | - Belinda O Darkwah
- Department of Pharmaceutical Sciences, School of Pharmacy, Cedarville University, Cedarville, OH, USA
| | - Stacy Lin
- Department of Pharmaceutical Sciences, School of Pharmacy, Cedarville University, Cedarville, OH, USA
| | - Andrea O Adegoke
- Department of Pharmaceutical Sciences, School of Pharmacy, Cedarville University, Cedarville, OH, USA
| | - Rebecca J Gryka
- Department of Pharmaceutical Sciences, School of Pharmacy, Cedarville University, Cedarville, OH, USA
| | - Denise S Jean-Louis
- Department of Pharmaceutical Sciences, School of Pharmacy, Cedarville University, Cedarville, OH, USA
| | - Samson Amos
- Department of Pharmaceutical Sciences, School of Pharmacy, Cedarville University, Cedarville, OH, USA
| |
Collapse
|
30
|
Maldonado MDM, Dharmawardhane S. Targeting Rac and Cdc42 GTPases in Cancer. Cancer Res 2018; 78:3101-3111. [PMID: 29858187 PMCID: PMC6004249 DOI: 10.1158/0008-5472.can-18-0619] [Citation(s) in RCA: 183] [Impact Index Per Article: 26.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Revised: 03/20/2018] [Accepted: 04/06/2018] [Indexed: 02/07/2023]
Abstract
Rac and Cdc42 are small GTPases that have been linked to multiple human cancers and are implicated in epithelial to mesenchymal transition, cell-cycle progression, migration/invasion, tumor growth, angiogenesis, and oncogenic transformation. With the exception of the P29S driver mutation in melanoma, Rac and Cdc42 are not generally mutated in cancer, but are overexpressed (gene amplification and mRNA upregulation) or hyperactivated. Rac and Cdc42 are hyperactivated via signaling through oncogenic cell surface receptors, such as growth factor receptors, which converge on the guanine nucleotide exchange factors that regulate their GDP/GTP exchange. Hence, targeting Rac and Cdc42 represents a promising strategy for precise cancer therapy, as well as for inhibition of bypass signaling that promotes resistance to cell surface receptor-targeted therapies. Therefore, an understanding of the regulatory mechanisms of these pivotal signaling intermediates is key for the development of effective inhibitors. In this review, we focus on the role of Rac and Cdc42 in cancer and summarize the regulatory mechanisms, inhibitory efficacy, and the anticancer potential of Rac- and Cdc42-targeting agents. Cancer Res; 78(12); 3101-11. ©2018 AACR.
Collapse
Affiliation(s)
- María Del Mar Maldonado
- Department of Biochemistry, School of Medicine, University of Puerto Rico, Medical Sciences Campus, San Juan, Puerto Rico
| | - Suranganie Dharmawardhane
- Department of Biochemistry, School of Medicine, University of Puerto Rico, Medical Sciences Campus, San Juan, Puerto Rico.
| |
Collapse
|
31
|
Luo Y, Shang P, Li D. Luteolin: A Flavonoid that Has Multiple Cardio-Protective Effects and Its Molecular Mechanisms. Front Pharmacol 2017; 8:692. [PMID: 29056912 PMCID: PMC5635727 DOI: 10.3389/fphar.2017.00692] [Citation(s) in RCA: 178] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2017] [Accepted: 09/19/2017] [Indexed: 01/14/2023] Open
Abstract
Cardiovascular disease (CVD) has become the leading cause of morbidity and mortality worldwide. A well-monitored diet with a sufficient intake of fruits and vegetables has been confirmed as a primary prevention of CVD. Plant constituents such as flavonoids have been shown to confer healthy benefits. Luteolin (Lut), a kind of flavonoid, possesses anti-oxidative, anti-tumor, and anti-inflammatory properties. Recent scientific literature has reported the cardiac protective effects of Lut in vitro and in vivo. Therefore, the aim of this review is to provide an update and detailed overview with cardio-protective molecular mechanisms of Lut with a focus on multiple intrinsic and extrinsic effectors. We further explore how these mechanisms participate in ischemia/reperfusion (I/R) injury, heart failure (HF) and atherosclerosis (AS). A proper understanding of the cardiovascular protective effects and the relative mechanisms of Lut may provide the possibility of new drug design and development for CVD. With the previous studies mainly focused on basic research, we need to advance the prospects of its further clinical utilization against CVD, large prospective clinical trials of Lut are needed to observe its therapeutic effects on patients with I/R injury, HF and AS, especially on the effective therapeutic dosage, and safety of long-term administration.
Collapse
Affiliation(s)
- Yuanyuan Luo
- The First Clinical College, Nanjing University of Chinese Medicine, Nanjing, China.,The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Pingping Shang
- Institute of Cardiovascular Disease Research, Xuzhou Medical University, Xuzhou, China
| | - Dongye Li
- The First Clinical College, Nanjing University of Chinese Medicine, Nanjing, China.,The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China.,Institute of Cardiovascular Disease Research, Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
32
|
Jackson S, ElAli A, Virgintino D, Gilbert MR. Blood-brain barrier pericyte importance in malignant gliomas: what we can learn from stroke and Alzheimer's disease. Neuro Oncol 2017; 19:1173-1182. [PMID: 28541444 PMCID: PMC5570196 DOI: 10.1093/neuonc/nox058] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The pericyte, a constitutive component of the central nervous system, is a poorly understood cell type that envelops the endothelial cell with the intended purpose of regulating vascular flow and endothelial cell permeability. Previous studies of pericyte function have been limited to a small number of disease processes such as ischemic stroke and Alzheimer's disease. Recently, publications have postulated a link between glioma stem cell differentiation and pericyte function. These studies suggest that there may be an important interaction of pericytes with tumor cells and other components of the tumor microenvironment in malignant primary glial neoplasms, most notably glioblastoma. This potential cellular interaction underscores the need to pursue more investigations of pericytes in malignant brain tumor biology. In this review, we summarize the functional roles of pericytes, particularly focusing on changes in pericyte biology during response to immune cells, inflammation, and hypoxic conditions. The information presented is based on the available data from studies of pericyte function in other central nervous system diseases but will serve as a foundation for research investigations to further understand the role of pericytes in malignant gliomas.
Collapse
Affiliation(s)
- Sadhana Jackson
- National Cancer Institute, Neuro-oncology Branch, Bethesda, Maryland; Research Center of CHU de Québec-Université Laval, Neuroscience Axis, Quebec, Canada; Department of Basic Medical Sciences, Neurosciences, and Sensory Organs, Human Anatomy and Histology Unit, University of Bari School of Medicine, Bari, Italy
| | - Ayman ElAli
- National Cancer Institute, Neuro-oncology Branch, Bethesda, Maryland; Research Center of CHU de Québec-Université Laval, Neuroscience Axis, Quebec, Canada; Department of Basic Medical Sciences, Neurosciences, and Sensory Organs, Human Anatomy and Histology Unit, University of Bari School of Medicine, Bari, Italy
| | - Daniela Virgintino
- National Cancer Institute, Neuro-oncology Branch, Bethesda, Maryland; Research Center of CHU de Québec-Université Laval, Neuroscience Axis, Quebec, Canada; Department of Basic Medical Sciences, Neurosciences, and Sensory Organs, Human Anatomy and Histology Unit, University of Bari School of Medicine, Bari, Italy
| | - Mark R Gilbert
- National Cancer Institute, Neuro-oncology Branch, Bethesda, Maryland; Research Center of CHU de Québec-Université Laval, Neuroscience Axis, Quebec, Canada; Department of Basic Medical Sciences, Neurosciences, and Sensory Organs, Human Anatomy and Histology Unit, University of Bari School of Medicine, Bari, Italy
| |
Collapse
|
33
|
Lefranc F, Tabanca N, Kiss R. Assessing the anticancer effects associated with food products and/or nutraceuticals using in vitro and in vivo preclinical development-related pharmacological tests. Semin Cancer Biol 2017; 46:14-32. [PMID: 28602819 DOI: 10.1016/j.semcancer.2017.06.004] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2017] [Revised: 06/02/2017] [Accepted: 06/02/2017] [Indexed: 10/19/2022]
Abstract
This review is part of a special issue entitled "Role of dietary pattern, foods, nutrients and nutraceuticals in supporting cancer prevention and treatment" and describes a pharmacological strategy to determine the potential contribution of food-related components as anticancer agents against established cancer. Therefore, this review does not relate to chemoprevention, which is analysed in several other reviews in the current special issue, but rather focuses on the following: i) the biological events that currently represent barriers against the treatment of certain types of cancers, primarily metastatic cancers; ii) the in vitro and in vivo pharmacological pre-clinical tests that can be used to analyse the potential anticancer effects of food-related components; and iii) several examples of food-related components with anticancer effects. This review does not represent a catalogue-based listing of food-related components with more or less anticancer activity. By contrast, this review proposes an original pharmacological strategy that researchers can use to analyse the potential anticancer activity of any food-related component-e.g., by considering the crucial characteristics of cancer biological aggressiveness. This review also highlights that cancer patients undergoing chemotherapy should restrict the use of "food complements" without supervision by a medical nutritionist. By contrast, an equilibrated diet that includes the food-related components listed herein would be beneficial for cancer patients who are not undergoing chemotherapy.
Collapse
Affiliation(s)
- Florence Lefranc
- Service de Neurochirurgie, Hôpital Erasme, Université Libre de Bruxelles, 808 route de Lennik, 1070 Brussels, Belgium.
| | - Nurhayat Tabanca
- U.S Department of Agriculture-Agricultural Research Service, Subtropical Horticulture Research Station,13601 Old Cutler Rd., Miami, FL 33158, USA.
| | - Robert Kiss
- Retired-formerly at the Fonds National de la Recherche Scientifique (FRS-FNRS, Brussels, Belgium), 5 rue d'Egmont, 1000 Brussels, Belgium.
| |
Collapse
|
34
|
de Paula LB, Primo FL, Pinto MR, Morais PC, Tedesco AC. Evaluation of a chloroaluminium phthalocyanine-loaded magnetic nanoemulsion as a drug delivery device to treat glioblastoma using hyperthermia and photodynamic therapy. RSC Adv 2017. [DOI: 10.1039/c6ra26105a] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The study describes the development of magnetic nanoemulsion loaded with citrate-coated maghemite nanoparticles and photosensitizer and the in vitro studies using cell lines while combining the use of hyperthermia and photodynamic therapy therapies.
Collapse
Affiliation(s)
- L. B. de Paula
- Department of Chemistry
- Center of Nanotechnology and Tissue Engineering – Photobiology and Photomedicine Research Group
- Faculty of Philosophy
- Science and Letters of Ribeirão Preto
- University of São Paulo
| | - F. L. Primo
- São Paulo State University (UNESP)
- School of Pharmaceutical Sciences
- Brazil
| | - M. R. Pinto
- Department of Chemistry
- Laboratory of Enzymology
- Faculty of Philosophy
- Science and Letters of Ribeirão Preto
- University of São Paulo
| | - P. C. Morais
- Institute of Physics
- University of Brasilia
- 70910-900 Brasília
- Brazil
- College of Chemistry and Chemical Engineering
| | - A. C. Tedesco
- Department of Chemistry
- Center of Nanotechnology and Tissue Engineering – Photobiology and Photomedicine Research Group
- Faculty of Philosophy
- Science and Letters of Ribeirão Preto
- University of São Paulo
| |
Collapse
|
35
|
Cook MT, Liang Y, Besch-Williford C, Hyder SM. Luteolin inhibits lung metastasis, cell migration, and viability of triple-negative breast cancer cells. BREAST CANCER-TARGETS AND THERAPY 2016; 9:9-19. [PMID: 28096694 PMCID: PMC5207335 DOI: 10.2147/bctt.s124860] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Most breast cancer-related deaths from triple-negative breast cancer (TNBC) occur following metastasis of cancer cells and development of tumors at secondary sites. Because TNBCs lack the three receptors targeted by current chemotherapeutic regimens, they are typically treated with extremely aggressive and highly toxic non-targeted treatment strategies. Women with TNBC frequently develop metastatic lesions originating from drug-resistant residual cells and have poor prognosis. For this reason, novel therapeutic strategies that are safer and more effective are sought. Luteolin (LU) is a naturally occurring, non-toxic plant compound that has proven effective against several types of cancer. With this in mind, we conducted in vivo and in vitro studies to determine whether LU might suppress metastasis of TNBC. In an in vivo mouse metastasis model, LU suppressed metastasis of human MDA-MB-435 and MDA-MB-231 (4175) LM2 TNBC cells to the lungs. In in vitro assays, LU inhibited cell migration and viability of MDA-MB-435 and MDA-MB-231 (4175) LM2 cells. Further, LU induced apoptosis in MDA-MB-231 (4175) LM2 cells. Relatively low levels (10 µM) of LU significantly inhibited vascular endothelial growth factor (VEGF) secretion in MDA-MB-231 (4175) LM2 cells, suggesting that it has the ability to suppress a potent angiogenic and cell survival factor. In addition, migration of MDA-MB-231 (4175) LM2 cells was inhibited upon exposure to an antibody against the VEGF receptor, KDR, but not by exposure to a VEGF165 antibody. Collectively, these data suggest that the anti-metastatic properties of LU may, in part, be due to its ability to block VEGF production and KDR-mediated activity, thereby inhibiting tumor cell migration. These studies suggest that LU deserves further investigation as a potential treatment option for women with TNBC.
Collapse
Affiliation(s)
- Matthew T Cook
- Department of Biomedical Sciences; Dalton Cardiovascular Research Center, University of Missouri
| | - Yayun Liang
- Department of Biomedical Sciences; Dalton Cardiovascular Research Center, University of Missouri
| | | | - Salman M Hyder
- Department of Biomedical Sciences; Dalton Cardiovascular Research Center, University of Missouri
| |
Collapse
|
36
|
Lestanova Z, Puerta F, Alanazi M, Bacova Z, Kiss A, Castejon AM, Bakos J. Downregulation of Oxytocin Receptor Decreases the Length of Projections Stimulated by Retinoic Acid in the U-87MG Cells. Neurochem Res 2016; 42:1006-1014. [DOI: 10.1007/s11064-016-2133-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Revised: 11/03/2016] [Accepted: 12/03/2016] [Indexed: 12/24/2022]
|
37
|
Abstract
Many food-derived phytochemical compounds and their derivatives represent a cornucopia of new anticancer compounds. Despite extensive study of luteolin, the literature has no information on the exact mechanisms or molecular targets through which it deters cancer progression. This review discusses existing data on luteolin's anticancer activities and then offers possible explanations for and molecular targets of its cancer-preventive action. Luteolin prevents tumor development largely by inactivating several signals and transcription pathways essential for cancer cells. This review also offers insights into the molecular mechanisms and targets through which luteolin either prevents cancer or mediates cancer cell death.
Collapse
|
38
|
Liu X, Yao Z. Chronic over-nutrition and dysregulation of GSK3 in diseases. Nutr Metab (Lond) 2016; 13:49. [PMID: 27493677 PMCID: PMC4972972 DOI: 10.1186/s12986-016-0108-8] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Accepted: 07/21/2016] [Indexed: 12/16/2022] Open
Abstract
Loss of cellular response to hormonal regulation in maintaining metabolic homeostasis is common in the process of aging. Chronic over-nutrition may render cells insensitive to such a hormonal regulation owing to overstimulation of certain signaling pathways, thus accelerating aging and causing diseases. The glycogen synthase kinase 3 (GSK3) plays a pivotal role in relaying various extracellular and intracellular regulatory signals critical to cell growth, survival, regeneration, or death. The main signaling pathway regulating GSK3 activity through serine-phosphorylation is the phosphoinositide 3-kinase (PI3K)/phosphoinositide-dependent kinase-1 (PDK1)/Akt relay that catalyzes serine-phosphorylation and thus inactivation of GSK3. In addition, perilipin 2 (PLIN2) has recently been shown to regulate GSK3 activation through direct association with GSK3. This review summarizes current understanding on environmental and nutritional factors contributing to GSK3 regulation (or dysregulation) through the PI3K/PDK1/Akt/GSK3 axis, and highlights the newly discovered role that PLIN2 plays in regulating GSK3 activity and GSK3 downstream pathways.
Collapse
Affiliation(s)
- Xunxian Liu
- Department of Biochemistry, Microbiology and Immunology, Ottawa Institute of Systems Biology, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5 Canada
| | - Zemin Yao
- Department of Biochemistry, Microbiology and Immunology, Ottawa Institute of Systems Biology, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5 Canada
| |
Collapse
|
39
|
Lin CM, Lin YL, Ho SY, Chen PR, Tsai YH, Chung CH, Hwang CH, Tsai NM, Tzou SC, Ke CY, Chang J, Chan YL, Wang YS, Chi KH, Liao KW. The inhibitory effect of 7,7″-dimethoxyagastisflavone on the metastasis of melanoma cells via the suppression of F-actin polymerization. Oncotarget 2016; 8:60046-60059. [PMID: 28947953 PMCID: PMC5601121 DOI: 10.18632/oncotarget.10960] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Accepted: 06/29/2016] [Indexed: 11/25/2022] Open
Abstract
7,7″-Dimethoxyagastisflavone (DMGF), a biflavonoid isolated from Taxus × media cv. Hicksii, induces apoptotic and autophagic cell death. However, whether DMGF suppresses tumor metastasis is unclear. The aim of this study was to investigate the anti-metastatic activities of DMGF on the metastatic processes of melanoma cells in vivo and in vitro. A transwell assay showed that DMGF could effectively attenuate the motility of B16F10 cells, and the results of real-time PCR revealed that DMGF also suppressed the expressions of matrix metalloproteinase-2 (MMP-2). Moreover, DMGF did not influence tube formation but inhibited the migration of endothelial cells. Furthermore, animal models were used to monitor the effects of DMGF on tumor metastasis, and all models showed that DMGF significantly suppressed the metastatic behaviors of B16F10 cells, including intravasation, colonization, and invasion of the lymphatic duct. In addition, DMGF could also reduce the densities of the blood vessels in the tumor area in vivo. Further investigation of the molecular mechanisms of anti-metastatic activity revealed that DMGF can down-regulate the levels of key modulators of the Cdc42/Rac1 pathway to interfere in F-actin polymerization and suppress the formation of lamellipodia by reducing the phosphorylation of CREB. These data suggested that DMGF presents anti-metastatic activities in B16F10 melanoma cells. Here, we demonstrated that DMGF can inhibit the metastasis of highly invasive melanoma cancer cells through the down-regulation of F-actin polymerization. Considering these findings, DMGF may be further developed to serve as a chemoprevention drug for patients with metastatic melanoma.
Collapse
Affiliation(s)
- Ching-Min Lin
- Institute of Molecular Medicine and Bioengineering, National Chiao Tung University, Hsinchu, Taiwan
| | - Yu-Ling Lin
- Center for Bioinformatics Research, National Chiao Tung University, Hsinchu, Taiwan.,Department of Biological Science and Technology, National Chiao Tung University, Hsinchu, Taiwan
| | - Shu-Yi Ho
- Department of Biological Science and Technology, National Chiao Tung University, Hsinchu, Taiwan
| | - Pin-Rong Chen
- Institute of Molecular Medicine and Bioengineering, National Chiao Tung University, Hsinchu, Taiwan
| | - Yi-Hsuan Tsai
- Institute of Molecular Medicine and Bioengineering, National Chiao Tung University, Hsinchu, Taiwan
| | - Chen-Han Chung
- Institute of Molecular Medicine and Bioengineering, National Chiao Tung University, Hsinchu, Taiwan
| | | | - Nu-Man Tsai
- Department of Medical and Laboratory Biotechnology, Chung Shan Medical University, Taichung, Taiwan.,Clinical Laboratory, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Shey-Cherng Tzou
- Institute of Molecular Medicine and Bioengineering, National Chiao Tung University, Hsinchu, Taiwan.,Department of Biological Science and Technology, National Chiao Tung University, Hsinchu, Taiwan
| | - Chun-Yen Ke
- Institute of Molecular Medicine and Bioengineering, National Chiao Tung University, Hsinchu, Taiwan
| | - Jung Chang
- Department of Biological Science and Technology, National Chiao Tung University, Hsinchu, Taiwan
| | - Yi-Lin Chan
- Department of Life Science, Chinese Culture University, Taichung, Taiwan
| | - Yu-Shan Wang
- Department of Radiation Therapy and Oncology, Shin Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan
| | - Kwan-Hwa Chi
- Department of Radiation Therapy and Oncology, Shin Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan
| | - Kuang-Wen Liao
- Institute of Molecular Medicine and Bioengineering, National Chiao Tung University, Hsinchu, Taiwan.,Department of Biological Science and Technology, National Chiao Tung University, Hsinchu, Taiwan.,Graduate Institut of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| |
Collapse
|
40
|
Meng G, Chai K, Li X, Zhu Y, Huang W. Luteolin exerts pro-apoptotic effect and anti-migration effects on A549 lung adenocarcinoma cells through the activation of MEK/ERK signaling pathway. Chem Biol Interact 2016; 257:26-34. [PMID: 27474067 DOI: 10.1016/j.cbi.2016.07.028] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Revised: 07/03/2016] [Accepted: 07/24/2016] [Indexed: 11/17/2022]
Abstract
An increasing amount of evidence suggests that luteolin, a common dietary flavonoid that is widely distributed in plants and foods, has been shown to be protective against cancer. However, the precise underlying mechanisms of its action against lung cancer are still poorly understood. In the present study, we investigated whether luteolin exhibits the anti-cancer effect in lung cancer through the induction of cell apoptosis and inhibition of cell migration, and whether mitogen-activated protein kinases (MAPKs) and Akt signaling pathways are required. Results revealed that luteolin exerted an anti-proliferation effect in a dose- and time-dependent manner in A549 lung adenocarcinoma cells, and induced apoptosis with a concomitant increase in the activation of caspases-3 and -9, diminution of Bcl-2, elevation in Bax expression, and the phosphorylation of MEK and its down-stream kinase ERK, as well as the activation of Akt. Luteolin also dramatically inhibited cell motility and migration in A549 cells. The inhibitor of MEK-ERK pathway protected against luteolin-induced cell death and suppressed the apoptosis-inducing and anti-migratory effects of luteolin, suggesting MEK-ERK signaling pathway plays an important role in mediating the pro-apoptotic effect and anti-migration effects of luteolin. Taken together, this study provides a new insight into the mode of action of luteolin on lung cancer.
Collapse
Affiliation(s)
- Guanmin Meng
- Department of Clinical Laboratory, Tongde Hospital of Zhejiang Province, Hangzhou, China; Department of Biochemistry, University of Alberta, Edmonton, Alberta, Canada
| | - Kequn Chai
- Department of Oncology, Tongde Hospital of Zhejiang Province, Hangzhou, China
| | - Xinda Li
- Department of Chemical and Materials Engineering, University of Alberta, Edmonton, Alberta, Canada
| | - Yongqiang Zhu
- Zhejiang Academy of Traditional Chinese Medicine, Tongde Hospital of Zhejiang Province, Hangzhou, China
| | - Weihua Huang
- Zhejiang Academy of Traditional Chinese Medicine, Tongde Hospital of Zhejiang Province, Hangzhou, China.
| |
Collapse
|
41
|
Poudel B, Ki HH, Luyen BTT, Lee YM, Kim YH, Kim DK. Triticumoside induces apoptosis via caspase-dependent mitochondrial pathway and inhibits migration through downregulation of MMP2/9 in human lung cancer cells. Acta Biochim Biophys Sin (Shanghai) 2016; 48:153-60. [PMID: 26758192 DOI: 10.1093/abbs/gmv124] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Accepted: 10/08/2015] [Indexed: 12/26/2022] Open
Abstract
Non-small cell lung cancer (NSCLC) is the major cancer-related death worldwide with only 14% five-year survival rate. Triticumoside, a phenolic compound present in Triticum aestivum sprout extract, has been recognized to have antiobesity and anti-inflammatory effects. However, the effect of triticumoside on cancer cell proliferation and migration has not been studied. In order to elucidate whether triticumoside exhibits an anticancer effect, cells were incubated with different doses of triticumoside, and apoptosis was assessed by observing cell viability, cellular morphological changes, and annexin-V-fluorescein isothiocyanate/propidium iodide staining. Cell cycle analysis, western blotting, wound healing assay, and quantitative-polymerase chain reaction were also performed. Triticumoside exhibited marked cytotoxicity in the cells in dose- and time-dependent manner. Triticumoside caused morphological changes, including cellular rounding, nuclear condensation, and shrinkage. Likewise, triticumoside enhanced the sub-G1 proportion of cells. Additionally, triticumoside regulated expression of apoptosis-associated proteins, such as B-cell lymphoma 2 (Bcl-2), Bcl-2-associated X, and procaspase-3/9. Triticumoside also inhibited migration of the cells through downregulation of matrix metalloproteinase-2/9 (MMP2/9). Collectively, these results suggest that triticumoside induces apoptosis through caspase-dependent mitochondrial pathway and suppresses migration via inhibition of MMP2/9 in NSCLC A549 cells.
Collapse
Affiliation(s)
- Barun Poudel
- Department of Immunology and Institute for Medical Sciences, Chonbuk National University Medical School, Jeonju, Jeonbuk 561-756, Korea
| | - Hyeon-Hui Ki
- Department of Immunology and Institute for Medical Sciences, Chonbuk National University Medical School, Jeonju, Jeonbuk 561-756, Korea
| | - Bui Thi Thuy Luyen
- College of Pharmacy, Chungnam National University, Daejeon 305-764, Korea
| | - Young-Mi Lee
- Department of Oriental Pharmacy, College of Pharmacy and Wonkwang-Oriental Medicines Research Institute, Wonkwang University, Iksan, Jeonbuk 570-749, Korea
| | - Young-Ho Kim
- College of Pharmacy, Chungnam National University, Daejeon 305-764, Korea
| | - Dae-Ki Kim
- Department of Immunology and Institute for Medical Sciences, Chonbuk National University Medical School, Jeonju, Jeonbuk 561-756, Korea
| |
Collapse
|
42
|
Chakrabarti M, Ray SK. Anti-tumor activities of luteolin and silibinin in glioblastoma cells: overexpression of miR-7-1-3p augmented luteolin and silibinin to inhibit autophagy and induce apoptosis in glioblastoma in vivo. Apoptosis 2015; 21:312-28. [DOI: 10.1007/s10495-015-1198-x] [Citation(s) in RCA: 93] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
43
|
Yu SH, Zhang CL, Dong FS, Zhang YM. miR-99a suppresses the metastasis of human non-small cell lung cancer cells by targeting AKT1 signaling pathway. J Cell Biochem 2015; 116:268-76. [PMID: 25187230 DOI: 10.1002/jcb.24965] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2014] [Accepted: 08/29/2014] [Indexed: 02/06/2023]
Abstract
MicroRNAs (miRNAs) play an important role in the development and progression of non-small cell lung cancer (NSCLC). Recently, several studies have shown that miR-99a is downregulated in various cancers, which can affect tumor initiation and maintenance. Herein, we found that miR-99a was downregulated in NSCLC tissues and suppressed tumor metastasis of NSCLC cells. Down-regulation of miR-99a is significantly associated with last-stage and tumor metastasis in NSCLC patients. Further functional experiments found that overexpression of miR-99a inhibit cell proliferation, migration, and invasion of NSCLC cells in vitro and tumor metastasis of NSCLC in vivo. In addition, we also found that AKT1 is directly involved in miR-99a-mediated tumor suppression. Restored the expression of AKT1 partially abolished the suppressive effects miR-99a on proliferation and invasion of NSCLC cells. Collectively, our data suggest that miR-99a plays an important role in the tumorigenesis and metastasis of NSCLC and may serve as a therapeutic target to avoid dissemination of NSCLC cells.
Collapse
Affiliation(s)
- Shi-huan Yu
- Department of Pulmonary Disease, First Affiliated Hospital of Harbin Medical University, Harbin, Helongjiang, 150001, P.R. China
| | | | | | | |
Collapse
|
44
|
Zeniou M, Fève M, Mameri S, Dong J, Salomé C, Chen W, El-Habr EA, Bousson F, Sy M, Obszynski J, Boh A, Villa P, Assad Kahn S, Didier B, Bagnard D, Junier MP, Chneiweiss H, Haiech J, Hibert M, Kilhoffer MC. Chemical Library Screening and Structure-Function Relationship Studies Identify Bisacodyl as a Potent and Selective Cytotoxic Agent Towards Quiescent Human Glioblastoma Tumor Stem-Like Cells. PLoS One 2015; 10:e0134793. [PMID: 26270679 PMCID: PMC4536076 DOI: 10.1371/journal.pone.0134793] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Accepted: 07/14/2015] [Indexed: 01/11/2023] Open
Abstract
Cancer stem-like cells reside in hypoxic and slightly acidic tumor niches. Such microenvironments favor more aggressive undifferentiated phenotypes and a slow growing "quiescent state" which preserves them from chemotherapeutic agents that essentially target proliferating cells. Our objective was to identify compounds active on glioblastoma stem-like cells, including under conditions that mimick those found in vivo within this most severe and incurable form of brain malignancy. We screened the Prestwick Library to identify cytotoxic compounds towards glioblastoma stem-like cells, either in a proliferating state or in more slow-growing "quiescent" phenotype resulting from non-renewal of the culture medium in vitro. Compound effects were assessed by ATP-level determination using a cell-based assay. Twenty active molecules belonging to different pharmacological classes have thus been identified. Among those, the stimulant laxative drug bisacodyl was the sole to inhibit in a potent and specific manner the survival of quiescent glioblastoma stem-like cells. Subsequent structure-function relationship studies led to identification of 4,4'-dihydroxydiphenyl-2-pyridyl-methane (DDPM), the deacetylated form of bisacodyl, as the pharmacophore. To our knowledge, bisacodyl is currently the only known compound targeting glioblastoma cancer stem-like cells in their quiescent, more resistant state. Due to its known non-toxicity in humans, bisacodyl appears as a new potential anti-tumor agent that may, in association with classical chemotherapeutic compounds, participate in tumor eradication.
Collapse
Affiliation(s)
- Maria Zeniou
- Laboratoire d’Innovation Thérapeutique, Université de Strasbourg / CNRS UMR7200, Laboratoire d’Excellence Medalis, Faculté de Pharmacie, 74 route du Rhin, 67401 Illkirch, France
- * E-mail:
| | - Marie Fève
- Laboratoire d’Innovation Thérapeutique, Université de Strasbourg / CNRS UMR7200, Laboratoire d’Excellence Medalis, Faculté de Pharmacie, 74 route du Rhin, 67401 Illkirch, France
| | - Samir Mameri
- Laboratoire d’Innovation Thérapeutique, Université de Strasbourg / CNRS UMR7200, Laboratoire d’Excellence Medalis, Faculté de Pharmacie, 74 route du Rhin, 67401 Illkirch, France
| | - Jihu Dong
- Laboratoire d’Innovation Thérapeutique, Université de Strasbourg / CNRS UMR7200, Laboratoire d’Excellence Medalis, Faculté de Pharmacie, 74 route du Rhin, 67401 Illkirch, France
| | - Christophe Salomé
- Laboratoire d’Innovation Thérapeutique, Université de Strasbourg / CNRS UMR7200, Laboratoire d’Excellence Medalis, Faculté de Pharmacie, 74 route du Rhin, 67401 Illkirch, France
| | - Wanyin Chen
- Laboratoire d’Innovation Thérapeutique, Université de Strasbourg / CNRS UMR7200, Laboratoire d’Excellence Medalis, Faculté de Pharmacie, 74 route du Rhin, 67401 Illkirch, France
| | - Elias A. El-Habr
- Neuroscience Paris Seine-IBPS, CNRS UMR 8246/ Inserm U1130/ UPMC UMCR18, 7 quai Saint Bernard, 75005 Paris, France
| | - Fanny Bousson
- Laboratoire d’Innovation Thérapeutique, Université de Strasbourg / CNRS UMR7200, Laboratoire d’Excellence Medalis, Faculté de Pharmacie, 74 route du Rhin, 67401 Illkirch, France
| | - Mohamadou Sy
- Laboratoire d’Innovation Thérapeutique, Université de Strasbourg / CNRS UMR7200, Laboratoire d’Excellence Medalis, Faculté de Pharmacie, 74 route du Rhin, 67401 Illkirch, France
| | - Julie Obszynski
- Laboratoire d’Innovation Thérapeutique, Université de Strasbourg / CNRS UMR7200, Laboratoire d’Excellence Medalis, Faculté de Pharmacie, 74 route du Rhin, 67401 Illkirch, France
| | - Alexandre Boh
- Laboratoire d’Innovation Thérapeutique, Université de Strasbourg / CNRS UMR7200, Laboratoire d’Excellence Medalis, Faculté de Pharmacie, 74 route du Rhin, 67401 Illkirch, France
| | - Pascal Villa
- Plateforme de Chimie Biologie Intégrative (PCBIS), Université de Strasbourg / CNRS UMS 3286, Laboratoire d’Excellence Medalis, ESBS Pôle API-Bld Sébastien Brant, 67401 Illkirch, France
| | - Suzana Assad Kahn
- Neuroscience Paris Seine-IBPS, CNRS UMR 8246/ Inserm U1130/ UPMC UMCR18, 7 quai Saint Bernard, 75005 Paris, France
| | - Bruno Didier
- Laboratoire d’Innovation Thérapeutique, Université de Strasbourg / CNRS UMR7200, Laboratoire d’Excellence Medalis, Faculté de Pharmacie, 74 route du Rhin, 67401 Illkirch, France
- Plateforme de Chimie Biologie Intégrative (PCBIS), Université de Strasbourg / CNRS UMS 3286, Laboratoire d’Excellence Medalis, ESBS Pôle API-Bld Sébastien Brant, 67401 Illkirch, France
| | - Dominique Bagnard
- U682, Inserm, Université de Strasbourg, 3, Avenue Molière, 67200 Strasbourg, France
| | - Marie-Pierre Junier
- Neuroscience Paris Seine-IBPS, CNRS UMR 8246/ Inserm U1130/ UPMC UMCR18, 7 quai Saint Bernard, 75005 Paris, France
| | - Hervé Chneiweiss
- Neuroscience Paris Seine-IBPS, CNRS UMR 8246/ Inserm U1130/ UPMC UMCR18, 7 quai Saint Bernard, 75005 Paris, France
| | - Jacques Haiech
- Laboratoire d’Innovation Thérapeutique, Université de Strasbourg / CNRS UMR7200, Laboratoire d’Excellence Medalis, Faculté de Pharmacie, 74 route du Rhin, 67401 Illkirch, France
| | - Marcel Hibert
- Laboratoire d’Innovation Thérapeutique, Université de Strasbourg / CNRS UMR7200, Laboratoire d’Excellence Medalis, Faculté de Pharmacie, 74 route du Rhin, 67401 Illkirch, France
| | - Marie-Claude Kilhoffer
- Laboratoire d’Innovation Thérapeutique, Université de Strasbourg / CNRS UMR7200, Laboratoire d’Excellence Medalis, Faculté de Pharmacie, 74 route du Rhin, 67401 Illkirch, France
| |
Collapse
|
45
|
Nash LA, Ward WE. Tea and bone health: Findings from human studies, potential mechanisms, and identification of knowledge gaps. Crit Rev Food Sci Nutr 2015; 57:1603-1617. [DOI: 10.1080/10408398.2014.1001019] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
46
|
Barrette K, Van Kelst S, Wouters J, Marasigan V, Fieuws S, Agostinis P, Oord J, Garmyn M. Epithelial‐mesenchymal transition during invasion of cutaneous squamous cell carcinoma is paralleled by
AKT
activation. Br J Dermatol 2014; 171:1014-21. [DOI: 10.1111/bjd.12967] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/10/2014] [Indexed: 12/31/2022]
Affiliation(s)
- K. Barrette
- Laboratory of Dermatology Department of Oncology KU Leuven Leuven Belgium
| | - S. Van Kelst
- Laboratory of Dermatology Department of Oncology KU Leuven Leuven Belgium
| | - J. Wouters
- Translational Cell & Tissue Research Department of Imaging and Pathology KU Leuven Leuven Belgium
| | - V. Marasigan
- Laboratory of Dermatology Department of Oncology KU Leuven Leuven Belgium
| | - S. Fieuws
- I‐Biostat Interuniversity Institute for Biostatistics and Statistical Bioinformatics KU Leuven and Universiteit Hasselt Leuven Belgium
| | - P. Agostinis
- Laboratory of Cell Death Research and Therapy Department of Cellular and Molecular Medicine KU Leuven Leuven Belgium
| | - J. Oord
- Translational Cell & Tissue Research Department of Imaging and Pathology KU Leuven Leuven Belgium
| | - M. Garmyn
- Laboratory of Dermatology Department of Oncology KU Leuven Leuven Belgium
| |
Collapse
|
47
|
Ebaid H. Promotion of immune and glycaemic functions in streptozotocin-induced diabetic rats treated with un-denatured camel milk whey proteins. Nutr Metab (Lond) 2014; 11:31. [PMID: 25009576 PMCID: PMC4087124 DOI: 10.1186/1743-7075-11-31] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2014] [Accepted: 06/25/2014] [Indexed: 12/29/2022] Open
Abstract
T cell mediated autoimmune diabetes is characterized by immune cell infiltration of pancreatic islets and destruction of insulin-producing β-cells. This study was designed to assess the effect of whey proteins (WP) on the responsiveness of lymphocytes in rats after four months of Streptozotocin (STZ)-induced Type 1 diabetes (T1D). A diabetic group was supplemented with WP daily for five weeks at a dose of 100 mg/kg. Ribonucleic acid (RNA) was extracted from stimulated lymphocytes in order to analyse gene expressions using real time PCR and RT-PCR. PCR results were confirmed with ELISA. The proliferation capacity of lymphocytes and their homing to the spleen were studied. Antigen-activated lymphocytes showed that diabetes impaired the mRNA expression of the protein kinase B (Akt1), Cdc42, and the co-stimulatory molecule, CD28, which are important for cell survival, actin polymerization and T cell activation, respectively. Accordingly, proliferation of lymphocytes was found to be suppressed in diabetic rats, both in vivo and in vitro. WP was found to restore Akt1, Cdc42 and CD28 mRNA expression during diabetes to normal levels. WP, therefore, served to activate the proliferation of B lymphocytes in diabetic rats both in vivo and in vitro. Although WP was found to up-regulate mRNA expression of both interleukin (IL)-2 and interferon gamma (IFN-γ), it suppressed the proliferation activity of almost all T cell subsets. This was confirmed by WP normalizing the structure and function of ß cells. Meanwhile, WP was found to down regulate the mRNA expression of Tumor necrosis factor-alpha (TNF-α) and its programmed cell death-receptor (Fas). Taken together, the results of this study provide evidence for the potential impact of WP in the treatment of immune impairment in T1D, suggesting that it serves to reverse autoimmunity by suppressing autoreactive T cells and down regulating TNF-α and Fas, resulting in improved pancreatic ß cell structure and function.
Collapse
Affiliation(s)
- Hossam Ebaid
- Department of Zoology, College of Science, King Saud University, Saudi Arabia, Riyadh, KSA ; Department of Zoology, Faculty of Science, El-Minia University, El-Minia, Egypt
| |
Collapse
|
48
|
Charoenrungruang S, Chanvorachote P, Sritularak B, Pongrakhananon V. Gigantol, a bibenzyl from Dendrobium draconis, inhibits the migratory behavior of non-small cell lung cancer cells. JOURNAL OF NATURAL PRODUCTS 2014; 77:1359-66. [PMID: 24844664 DOI: 10.1021/np500015v] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
Lung cancer is one of the most common causes of cancer death due to its high metastasis potential. The process of cancer migration is an early step that is required for successful metastasis. The discovery and development of natural compounds for cancer therapy have garnered increasing attention in recent years. Gigantol (1) is a bibenzyl compound derived from the Thai orchid, Dendrobium draconis. It exhibits significant cytotoxic activity against several cancer cell lines; however, until recently, the role of 1 on tumor metastasis has not been characterized. This study demonstrates that 1 suppresses the migratory behavior of non-small cell lung cancer H460 cells. Western blot analysis reveals that 1 down-regulates caveolin-1 (Cav-1), activates ATP-dependent tyrosine kinase (phosphorylated Akt at Ser 473), and cell division cycle 42 (Cdc42), thereby suppressing filopodia formation. The inhibitory effect of 1 on cell movement is also exhibited in another lung cancer cell line, H292, but not in normal human keratinocytes (HaCat). The inhibitory activity of 1 on lung cancer migration suggests that this compound may be suitable for further development for the treatment of cancer metastasis.
Collapse
Affiliation(s)
- Sopanya Charoenrungruang
- Department of Pharmacology and Physiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University , Bangkok 10330, Thailand
| | | | | | | |
Collapse
|
49
|
Anti-hepatoma cells function of luteolin through inducing apoptosis and cell cycle arrest. Tumour Biol 2013; 35:3053-60. [DOI: 10.1007/s13277-013-1396-5] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2013] [Accepted: 11/05/2013] [Indexed: 12/24/2022] Open
|