1
|
Aebisher D, Woźnicki P, Czarnecka-Czapczyńska M, Dynarowicz K, Szliszka E, Kawczyk-Krupka A, Bartusik-Aebisher D. Molecular Determinants for Photodynamic Therapy Resistance and Improved Photosensitizer Delivery in Glioma. Int J Mol Sci 2024; 25:8708. [PMID: 39201395 PMCID: PMC11354549 DOI: 10.3390/ijms25168708] [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: 05/18/2024] [Revised: 08/02/2024] [Accepted: 08/06/2024] [Indexed: 09/02/2024] Open
Abstract
Gliomas account for 24% of all the primary brain and Central Nervous System (CNS) tumors. These tumors are diverse in cellular origin, genetic profile, and morphology but collectively have one of the most dismal prognoses of all cancers. Work is constantly underway to discover a new effective form of glioma therapy. Photodynamic therapy (PDT) may be one of them. It involves the local or systemic application of a photosensitive compound-a photosensitizer (PS)-which accumulates in the affected tissues. Photosensitizer molecules absorb light of the appropriate wavelength, initiating the activation processes leading to the formation of reactive oxygen species and the selective destruction of inappropriate cells. Research focusing on the effective use of PDT in glioma therapy is already underway with promising results. In our work, we provide detailed insights into the molecular changes in glioma after photodynamic therapy. We describe a number of molecules that may contribute to the resistance of glioma cells to PDT, such as the adenosine triphosphate (ATP)-binding cassette efflux transporter G2, glutathione, ferrochelatase, heme oxygenase, and hypoxia-inducible factor 1. We identify molecular targets that can be used to improve the photosensitizer delivery to glioma cells, such as the epithelial growth factor receptor, neuropilin-1, low-density lipoprotein receptor, and neuropeptide Y receptors. We note that PDT can increase the expression of some molecules that reduce the effectiveness of therapy, such as Vascular endothelial growth factor (VEGF), glutamate, and nitric oxide. However, the scientific literature lacks clear data on the effects of PDT on many of the molecules described, and the available reports are often contradictory. In our work, we highlight the gaps in this knowledge and point to directions for further research that may enhance the efficacy of PDT in the treatment of glioma.
Collapse
Affiliation(s)
- David Aebisher
- Department of Photomedicine and Physical Chemistry, Medical College of The Rzeszów University, 35-310 Rzeszów, Poland
| | - Paweł Woźnicki
- English Division Science Club, Medical College of The Rzeszów University, 35-310 Rzeszów, Poland;
| | - Magdalena Czarnecka-Czapczyńska
- Department of Internal Medicine, Angiology and Physical Medicine, Center for Laser Diagnostics and Therapy, Medical University of Silesia, Batorego 15 Street, 41-902 Bytom, Poland;
| | - Klaudia Dynarowicz
- Center for Innovative Research in Medical and Natural Sciences, Medical College of The University of Rzeszów, 35-310 Rzeszów, Poland;
| | - Ewelina Szliszka
- Department of Microbiology and Immunology, Medical University of Silesia, Poniatowskiego 15, 40-055 Katowice, Poland;
| | - Aleksandra Kawczyk-Krupka
- Department of Internal Medicine, Angiology and Physical Medicine, Center for Laser Diagnostics and Therapy, Medical University of Silesia, Batorego 15 Street, 41-902 Bytom, Poland;
| | - Dorota Bartusik-Aebisher
- Department of Biochemistry and General Chemistry, Medical College of The Rzeszów University, 35-310 Rzeszów, Poland;
| |
Collapse
|
2
|
Chen H, Zhu MZ, Wang XT, Ai M, Li SS, Wan MY, Wang PY, Cai WW, Hou B, Xu F, Lang F, Qiu LY, Zhou YT. 1,25(OH) 2 D 3 inhibits Lewis lung cancer cell migration via NHE1-sensitive metabolic reprograming. IUBMB Life 2024; 76:182-199. [PMID: 37921568 DOI: 10.1002/iub.2789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 09/28/2023] [Indexed: 11/04/2023]
Abstract
High prevalence and metastasis rates are characteristics of lung cancer. Glycolysis provides energy for the development and metastasis of cancer cells. The 1,25-dihydroxy vitamin D3 (1,25(OH)2 D3 ) has been linked to reducing cancer risk and regulates various physiological functions. We hypothesized that 1,25(OH)2 D3 could be associated with the expression and activity of Na+ /H+ exchanger isoform 1 (NHE1) of Lewis lung cancer cells, thus regulating glycolysis as well as migration by actin reorganization. Followed by online public data analysis, Vitamin D3 receptor, the receptor of 1,25(OH)2 D3 has been proved to be abundant in lung cancers. We demonstrated that 1,25(OH)2 D3 treatment suppressed transcript levels, protein levels, and activity of NHE1 in LLC cells. Furthermore, 1,25(OH)2 D3 treatment resets the metabolic balance between glycolysis and OXPHOS, mainly including reducing glycolytic enzymes expression and lactate production. In vivo experiments showed the inhibition effects on tumor growth as well. Therefore, we concluded that 1,25(OH)2 D3 could amend the NHE1 function, which leads to metabolic reprogramming and cytoskeleton reconstruction, finally inhibits the cell migration.
Collapse
Affiliation(s)
- Hong Chen
- Wuxi Medical School, Jiangnan University, Wuxi, People's Republic of China
| | - Mei-Zhen Zhu
- Wuxi Medical School, Jiangnan University, Wuxi, People's Republic of China
| | - Xi-Ting Wang
- Wuxi Medical School, Jiangnan University, Wuxi, People's Republic of China
| | - Min Ai
- Wuxi Medical School, Jiangnan University, Wuxi, People's Republic of China
- Laboratory Animal Center of Shanghai Jiao Tong University, Shanghai, People's Republic of China
| | - Shuang-Shuang Li
- Wuxi Medical School, Jiangnan University, Wuxi, People's Republic of China
- Shanghai Seventh People's Hospital, Shanghai, People's Republic of China
| | - Ming-Yu Wan
- Wuxi Medical School, Jiangnan University, Wuxi, People's Republic of China
| | - Pei-Yao Wang
- Wuxi Medical School, Jiangnan University, Wuxi, People's Republic of China
| | - Wei-Wei Cai
- Wuxi Medical School, Jiangnan University, Wuxi, People's Republic of China
| | - Bao Hou
- Wuxi Medical School, Jiangnan University, Wuxi, People's Republic of China
| | - Fei Xu
- Wuxi Medical School, Jiangnan University, Wuxi, People's Republic of China
| | - Florian Lang
- Department of Physiology, University of Tübingen, Tübingen, Germany
| | - Li-Ying Qiu
- Wuxi Medical School, Jiangnan University, Wuxi, People's Republic of China
| | - Yue-Tao Zhou
- Wuxi Medical School, Jiangnan University, Wuxi, People's Republic of China
| |
Collapse
|
3
|
Kang X, Chen J, Hou JF. HSP90 facilitates stemness and enhances glycolysis in glioma cells. BMC Neurol 2022; 22:420. [DOI: 10.1186/s12883-022-02924-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 10/10/2022] [Indexed: 11/13/2022] Open
Abstract
Abstract
Background
Glioma is one of the most commonly occurring malignant brain cancers with high recurrence and mortality. Glioma stem cells (SCs) are a rare sub-group of glioma cells that play a critical role in tumor progression. Heat shock protein 90 (HSP90) is known to promote the stemness of glioma SCs. Here, we investigated the role of HSP90 in glioma SC metabolism, to reveal its potential as a novel therapeutic target.
Methods
Self-renewal assays were used to assess stemness. Cell migration, invasion and viability were measured using Transwell and CCK-8 assays, respectively. Tumor growth was evaluated in xenograft nude mouse models. The expression of known markers of stemness including CD44, A2B5, Oct4, Nestin, Lgr5, Sox2, CD24 were assessed by western blotting. HSP90 expression was assessed by western blotting and immunohistochemistry (IHC). Glucose consumption, lactic acid production and ATP levels were measured using commercially available kits. Extracellular acidification rates (ECAR) were measured using the Seahorse XFe/XF analyzer.
Results
HSP90 was upregulated in spheroid cells compared to parental cells. HSP90 facilitated the characteristics of SCs through enhancing self-renewal capacity, glucose consumption, lactic acid production, total ATP, ECAR and glycolysis. 2-DG, an inhibitor of glycolysis, reduced HSP90 expression and inhibited the stemness of glioma cells.
Conclusions
We show that HSP90 accelerates stemness and enhances glycolysis in glioma cells. Inhibition of glycolysis with 2DG prevented stemness. This reveals new roles for HSP90 during glioma progression and highlights this protein as a potential target for much-needed anti-glioma therapeutics.
Collapse
|
4
|
Han Z, Mou Z, Jing Y, Jiang R, Sun T. EMX1 functions as a tumor inhibitor in spinal cord glioma through transcriptional suppression of WASF2 and inactivation of the Wnt/β-catenin axis. Brain Behav 2022; 12:e2684. [PMID: 35849030 PMCID: PMC9392518 DOI: 10.1002/brb3.2684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 06/12/2022] [Accepted: 06/14/2022] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND Gliomas are the most frequent and aggressive cancers in the central nervous system, and spinal cord glioma (SCG) is a rare class of the gliomas. Empty spiracles homobox genes (EMXs) have shown potential tumor suppressing roles in glioma, but the biological function of EMX1 in SCG is unclear. METHODS The EMX1 expression in clinical tissues of patients with SCG was examined. SCG cells were extracted from the tissues, and altered expression of EMX1 was then introduced to examine the role of EMX1 in cell growth and invasiveness in vitro. Xenograft tumors were induced in nude mice for in vivo validation. The targets of EXM1 were predicted via bioinformatic analysis and validated by luciferase and ChIP-qPCR assays. Rescue experiments were conducted to validate the involvements of the downstream molecules. RESULTS EMX1 was poorly expressed in glioma, which was linked to decreased survival rate of patients according to the bioinformatics prediction. In clinical tissues, EMX1 was poorly expressed in SCG, especially in the high-grade tissues. EMX1 upregulation significantly suppressed growth and metastasis of SCG cells in vitro and in vivo. EMX1 bound to the promoter of WASP family member 2 (WASF2) to suppress its transcription. Restoration of WASF2 blocked the tumor-suppressing effect of EMX1. EMX1 suppressed Wnt/β-catenin signaling activity by inhibiting WASF2. Coronaridine, a Wnt/β-catenin-specific antagonist, blocked SCG cell growth and metastasis induced by WASF2. CONCLUSION This study elucidates that EMX1 functions as a tumor inhibitor in SCG by suppressing WASF2-dependent activation of the Wnt/β-catenin axis.
Collapse
Affiliation(s)
- Ziyin Han
- Department of Traumatic Orthopedics, Yantaishan Hospital of Yantai, Yantai, Shandong, P.R. China
| | - Zufang Mou
- Administration Department of Nosocomial Infection, Yantaishan Hospital of Yantai, Yantai, Shandong, P.R. China
| | - Yulong Jing
- Department of Traumatic Orthopedics, Yantaishan Hospital of Yantai, Yantai, Shandong, P.R. China
| | - Rong Jiang
- Department of Physiology, Binzhou Medical University, Yantai Campus, Yantai, Shandong, P.R. China
| | - Tao Sun
- Department of Traumatic Orthopedics, Yantaishan Hospital of Yantai, Yantai, Shandong, P.R. China
| |
Collapse
|
5
|
Behnam M, Motamedzadeh A, Aalinezhad M, Dadgostar E, Rashidi Noshabad FZ, Pourfridoni M, Raei M, Mirzaei H, Aschner M, Tamtaji OR. The role of aquaporin 4 in brain tumors: implications for pathophysiology, diagnosis and therapy. Mol Biol Rep 2022; 49:10609-10615. [PMID: 35715607 DOI: 10.1007/s11033-022-07656-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 05/25/2022] [Accepted: 05/27/2022] [Indexed: 11/24/2022]
Abstract
Primary brain tumors are a heterogeneous group of tumors that arise from cells intrinsic to the central nervous system (CNS). Aquaporin-4 (AQP4) has been implicated in the pathogenesis of brain tumors. Previous reports have documented a relationship between AQP4 and several molecular pathways associated with the etiology of brain tumors, such as apoptosis, invasion and cell migration. AQP4 affects apoptosis via cytochrome C, Bad and Bcl-2, as well as invasion and migration via IDO1/TDO-Kyn-AhR axis, lncRNA LINC00461, miR-216a, miRNA-320a and MMPs. In addition, inhibition of AQP4 mitigates the progression of brain tumors. This review summarizes current knowledge and evidence regarding the relationship between AQP4 and brain tumors, and the mechanisms involved.
Collapse
Affiliation(s)
- Mohammad Behnam
- Student Research Committee, Kashan University of Medical Sciences, Kashan, I.R. of Iran
| | - Alireza Motamedzadeh
- Department of Internal Medicine, Faculty of Medicine, Kashan University of Medical Sciences, Kashan, I.R. of Iran
| | - Marzieh Aalinezhad
- Department of Radiology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, I.R. of Iran
| | - Ehsan Dadgostar
- Behavioral Sciences Research Center, Isfahan University of Medical Sciences, Isfahan, I.R. of Iran.,Student Research Committee, Isfahan University of Medical Sciences, Isfahan, I.R. of Iran
| | | | - Mohammad Pourfridoni
- Student Research Committee, Jiroft University of Medical Sciences, Jiroft, I.R. of Iran
| | - Maedeh Raei
- Faculty of Medicine, Sari Branch, Islamic Azad University, Sari, I.R. of Iran
| | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, I.R. of Iran.
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, 10461, Bronx, NY, USA
| | - Omid Reza Tamtaji
- Electrophysiology Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, I.R. of Iran. .,Department of Physiology, School of Medicine, Tehran University of Medical Sciences, Tehran, I.R. of Iran.
| |
Collapse
|
6
|
Nikolovska K, Seidler UE, Stock C. The Role of Plasma Membrane Sodium/Hydrogen Exchangers in Gastrointestinal Functions: Proliferation and Differentiation, Fluid/Electrolyte Transport and Barrier Integrity. Front Physiol 2022; 13:899286. [PMID: 35665228 PMCID: PMC9159811 DOI: 10.3389/fphys.2022.899286] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 04/19/2022] [Indexed: 12/11/2022] Open
Abstract
The five plasma membrane Na+/H+ exchanger (NHE) isoforms in the gastrointestinal tract are characterized by distinct cellular localization, tissue distribution, inhibitor sensitivities, and physiological regulation. NHE1 (Slc9a1) is ubiquitously expressed along the gastrointestinal tract in the basolateral membrane of enterocytes, but so far, an exclusive role for NHE1 in enterocyte physiology has remained elusive. NHE2 (Slc9a2) and NHE8 (Slc9a8) are apically expressed isoforms with ubiquitous distribution along the colonic crypt axis. They are involved in pHi regulation of intestinal epithelial cells. Combined use of a knockout mouse model, intestinal organoid technology, and specific inhibitors revealed previously unrecognized actions of NHE2 and NHE8 in enterocyte proliferation and differentiation. NHE3 (Slc9a3), expressed in the apical membrane of differentiated intestinal epithelial cells, functions as the predominant nutrient-independent Na+ absorptive mechanism in the gut. The new selective NHE3 inhibitor (Tenapanor) allowed discovery of novel pathophysiological and drug-targetable NHE3 functions in cystic-fibrosis associated intestinal obstructions. NHE4, expressed in the basolateral membrane of parietal cells, is essential for parietal cell integrity and acid secretory function, through its role in cell volume regulation. This review focuses on the expression, regulation and activity of the five plasma membrane Na+/H+ exchangers in the gastrointestinal tract, emphasizing their role in maintaining intestinal homeostasis, or their impact on disease pathogenesis. We point to major open questions in identifying NHE interacting partners in central cellular pathways and processes and the necessity of determining their physiological role in a system where their endogenous expression/activity is maintained, such as organoids derived from different parts of the gastrointestinal tract.
Collapse
|
7
|
Tiek D, Cheng SY. DNA damage and metabolic mechanisms of cancer drug resistance. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2022; 5:368-379. [PMID: 35800362 PMCID: PMC9255237 DOI: 10.20517/cdr.2021.148] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 02/11/2022] [Accepted: 03/11/2022] [Indexed: 11/13/2022]
Abstract
Cancer drug resistance is one of the main barriers to overcome to ensure durable treatment responses. While many pivotal advances have been made in first combination therapies, then targeted therapies, and now broadening out to immunomodulatory drugs or metabolic targeting compounds, drug resistance is still ultimately universally fatal. In this brief review, we will discuss different strategies that have been used to fight drug resistance from synthetic lethality to tumor microenvironment modulation, focusing on the DNA damage response and tumor metabolism both within tumor cells and their surrounding microenvironment. In this way, with a better understanding of both targetable mutations in combination with the metabolism, smarter drugs may be designed to combat cancer drug resistance.
Collapse
Affiliation(s)
- Deanna Tiek
- Correspondence to: Deanna Tiek, The Ken & Ruth Davee Department of Neurology, Lou and Jean Malnati Brain Tumor Institute at Northwestern Medicine, Robert H. Lurie Comprehensive Cancer Center, and Simpson Querry Institute for Epigenetics, Northwestern University, Feinberg School of Medicine, 303 E Superior St, Chicago, IL 60611, USA. E-mail: ; Shi-Yuan Cheng, The Ken & Ruth Davee Department of Neurology, Lou and Jean Malnati Brain Tumor Institute at Northwestern Medicine, Robert H. Lurie Comprehensive Cancer Center, and Simpson Querry Institute for Epigenetics, Northwestern University, Feinberg School of Medicine, 303 E Superior St, Chicago, IL 60611, USA. E-mail:
| | - Shi-Yuan Cheng
- Correspondence to: Deanna Tiek, The Ken & Ruth Davee Department of Neurology, Lou and Jean Malnati Brain Tumor Institute at Northwestern Medicine, Robert H. Lurie Comprehensive Cancer Center, and Simpson Querry Institute for Epigenetics, Northwestern University, Feinberg School of Medicine, 303 E Superior St, Chicago, IL 60611, USA. E-mail: ; Shi-Yuan Cheng, The Ken & Ruth Davee Department of Neurology, Lou and Jean Malnati Brain Tumor Institute at Northwestern Medicine, Robert H. Lurie Comprehensive Cancer Center, and Simpson Querry Institute for Epigenetics, Northwestern University, Feinberg School of Medicine, 303 E Superior St, Chicago, IL 60611, USA. E-mail:
| |
Collapse
|
8
|
Wang T, Xing Y, Cheng Z, Yu F. Analysis of Single Extracellular Vesicles for Biomedical Applications with Especial Emphasis on Cancer Investigations. Trends Analyt Chem 2022. [DOI: 10.1016/j.trac.2022.116604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
9
|
Wang X, Dong H. A convergent synthetic platform for anticancer drugs formulation with nanoparticle delivery for the treatment and nursing care of glioma cancer. Process Biochem 2021. [DOI: 10.1016/j.procbio.2021.10.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
10
|
Xie H, Lv S, Wang Z, Yuan X. E2F transcription factor 1 elevates cyclin D1 expression by suppressing transcription of microRNA-107 to augment progression of glioma. Brain Behav 2021; 11:e2399. [PMID: 34758200 PMCID: PMC8671784 DOI: 10.1002/brb3.2399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 09/29/2021] [Accepted: 09/30/2021] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND Dysregulation of microRNAs has been frequently implicated in the progression of human diseases, including glioma. This study aims to explore the interaction between E2F transcription factor 1 (E2F1) and miR-107 in the progression of glioma. METHODS Expression of miR-107 in glioma tissues and cells was examined. Putative binding sites between E2F1 and the promoter region of miR-107, and between miR-107 and cyclin D1 (CCND1) mRNA were predicted via bioinformatic systems and validated via chromatin immunoprecipitation and luciferase reporter gene assays. Altered expression of miR-107, E2F1, and CCND1 was introduced in A172 and T98G cells to examine their roles in cell growth and the activity of the Wnt/β-catenin signaling. In vivo experiments were performed by injecting cells in nude mice. RESULTS miR-107 was poorly expressed, whereas E2F1 and CCND1 were highly expressed in glioma tissues and cells. E2F1 bound to the promoter region of miR-107 to induce transcriptional repression, and miR-107 directly bound to CCND1 mRNA to reduce its expression. Overexpression of miR-107 reduced proliferation, migration and invasion, and augmented apoptosis of glioma cells, and it reduced activity of the Wnt/β-catenin pathway. The anti-tumorigenic roles of miR-107 were blocked by E2F1 or CCND1 overexpression. Similar results were reproduced in vivo where miR-107 overexpression or E2F1 inhibition blocked tumor growth in nude mice. CONCLUSION This study suggested that E2F1 reduces miR-107 transcription to induce CCND1 upregulation, which leads to progression of glioma via Wnt/β-catenin signaling activation.
Collapse
Affiliation(s)
- Huan Xie
- Department of Clinical Medicine, Jiangxi Health Vocational College, Nanchang, P. R. China
| | - Shigang Lv
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University, Nanchang, P. R. China
| | - Zhaozhen Wang
- Department of Clinical Medicine, Jiangxi Health Vocational College, Nanchang, P. R. China
| | - Xinzhang Yuan
- Department of Clinical Medicine, Jiangxi Health Vocational College, Nanchang, P. R. China
| |
Collapse
|
11
|
Na +/H +-Exchanger Family as Novel Prognostic Biomarkers in Colorectal Cancer. JOURNAL OF ONCOLOGY 2021; 2021:3241351. [PMID: 34759967 PMCID: PMC8575632 DOI: 10.1155/2021/3241351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/28/2021] [Accepted: 10/05/2021] [Indexed: 11/17/2022]
Abstract
Background The acidic characteristics of the tumor microenvironment (TME) are attributed to cancer cells' needs of metabolism which produce a large amount of H+. In order not to affect its own life activities, it needs to release H+ into the intercellular space through an efficient Na+/H+ exchanger. On account of the intestine whose physiological function is highly dependent on intestinal pH value, NHE family members may play a critical role in the occurrence and development of colorectal cancer (CRC). Methods TCGA, GEPIA2, ONCOMINE, UALCAN, STRING, TIMER, Cytoscape, TargetScan, ENCORI, LncBase v.2, DNMIVD, HPA, and CellMinerTM databases were used in our study. Results The mRNA expressions of SLC9A1, SLC9A2, SLC9A3, and SLC9A9 were evidently lower in COAD than in normal samples; however, the mRNA expressions of SLC9A5, SLC9A8, and SLC9B2 were higher. Besides, mRNA expressions of NHE family were extremely associated with clinicopathological features, tumor immune microenvironment and stemness score, DNA methylation, and patient prognosis in COAD. Moreover, we conjectured that NHE family may play a role through MAPK or ErbB signaling pathway according to the results of GO/KEGG enrichment analysis. At last, we found that NHE family members were key factors of various kinds of cancers. Conclusion Our study indicated that NHE family represented new diagnostic and therapeutic targets for CRC, which could have important significance for the clinical treatment of CRC.
Collapse
|
12
|
Tian W, Zhang Y, Liu H, Jin H, Sun T. LINC01123 potentially correlates with radioresistance in glioma through the miR-151a/CENPB axis. Neuropathology 2021; 42:3-15. [PMID: 34519373 DOI: 10.1111/neup.12764] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 05/17/2021] [Accepted: 05/24/2021] [Indexed: 11/29/2022]
Abstract
Radiotherapy represents the most effective nonsurgical therapy, whereas acquired radioresistance remains a major challenge in glioma treatment. Deregulation of long noncoding RNAs (lncRNAs) is frequently involved in tumorigenesis. This study investigates the role of LINC01123 in radioresistance in glioma with molecules involved. LINC01123 was identified as the most upregulated gene in a glioma gene expression dataset GSE103227. LINC01123 was highly expressed in the radioresistant glioma tissues radioresistant glioma U251 (U251R) cells. Downregulation of LINC01123 reduced cell proliferation and colony formation abilities, as well as resistance to apoptosis of the U251R cells after 4 Gy X-ray irradiation. The micro(mi)RNA-151a gene (miR-151a) was a poorly expressed miRNA in glioma, and it was a target of LINC01123. The centromere protein B gene (CENPB) mRNA was a direct target of miR-151a and demonstrated a positive correlation with LINC01123 in glioma tissues and cells. Further inhibition of miR-151a or overexpression of CENPB restored radioresistance of glioma cells. In addition, silencing of LINC01123 suppressed growth of xenograft tumors formed by U251R cells in nude mice. To conclude, the present study demonstrates that LINC01123 serves as a sponge for miR-151a and upregulates CENPB expression to increase the radioresistance of glioma cells in vitro and in vivo.
Collapse
Affiliation(s)
- Weicheng Tian
- Department of Radiotherapy, Jilin Cancer Hospital, Changchun, China
| | - Yingying Zhang
- Institution for Drug Clinical Trial, Jilin Cancer Hospital, Changchun, China
| | - Hairui Liu
- Department of Abdominal, Jilin Cancer Hospital, Changchun, China
| | - Haiguo Jin
- Department of Radiotherapy, Jilin Cancer Hospital, Changchun, China
| | - Tao Sun
- Department of Radiotherapy, Jilin Cancer Hospital, Changchun, China
| |
Collapse
|
13
|
Wang C, Tu H, Yang L, Ma C, Hu J, Luo J, Wang H. FOXN3 inhibits cell proliferation and invasion via modulating the AKT/MDM2/p53 axis in human glioma. Aging (Albany NY) 2021; 13:21587-21598. [PMID: 34511432 PMCID: PMC8457572 DOI: 10.18632/aging.203499] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Accepted: 07/30/2021] [Indexed: 12/15/2022]
Abstract
This study aimed to evaluate the biological role of forkhead box N3 (FOXN3) in human glioma and clarify the possible molecular mechanisms. FOXN3 expression patterns in clinical tissue specimens were characterized via qPCR and Western blotting. Kaplan-Meier survival curve was applied to assess the correlation between FOXN3 expression and overall survival. Effects of FOXN3 over-expression and depletion on glioma cell proliferation, apoptosis, migration and invasion were assessed by CCK8, colony formation assay, flow cytometry, scratch wound healing assay and Transwell invasion assay, respectively. Moreover, the involvement of AKT/murine double minute 2 (MDM2)/p53 pathway was evaluated. Additionally, tumor transplantation model assay was performed to determine the effects of FOXN3 over-expression on glioma cell growth in vivo. Results showed that FOXN3 was significantly down-regulated in glioma tissues compared with normal tissues. Patients with lower FOXN3 expression exhibited a shorter overall survival time. Gain- and loss-of-function analyses demonstrated that FOXN3 over-expression significantly suppressed proliferation, survival and motility of glioma cells, whereas FOXN3 knockdown remarkably promoted glioma cell proliferation, survival and motility. Furthermore, FOXN3 over-expression inhibited the activation of AKT/MDM2/p53 signaling pathway in glioma cells, while FOXN3 depletion facilitated its activation. Additionally, tumor xenograft assays revealed that FOXN3 over-expression retarded glioma cell growth in vivo. Collectively, these findings indicate that FOXN3 inhibits cell growth and invasion through inactivating the AKT/MDM2/p53 signaling pathway and that FOXN3-AKT/MDM2/p53 axis may represent a novel therapeutic target for glioma patients.
Collapse
Affiliation(s)
- Chaojia Wang
- Department of Neurosurgery, Taihe Affiliated Hospital of Hubei University of Medicine, Shiyan 442000, China
| | - Hanjun Tu
- First School of Clinical Medicine, Hubei University of Medicine, Shiyan 442000, China
| | - Ling Yang
- Department of Pediatrics, Taihe Affiliated Hospital of Hubei University of Medicine, Shiyan 442000, China
| | - Chunming Ma
- Department of Rehabilitation, Taihe Affiliated Hospital of Hubei University of Medicine, Shiyan 442000, China
| | - Juntao Hu
- Department of Neurosurgery, Taihe Affiliated Hospital of Hubei University of Medicine, Shiyan 442000, China
| | - Jie Luo
- Department of Neurosurgery, Taihe Affiliated Hospital of Hubei University of Medicine, Shiyan 442000, China
| | - Hui Wang
- Department of Neurosurgery, Taihe Affiliated Hospital of Hubei University of Medicine, Shiyan 442000, China
| |
Collapse
|
14
|
Yu Y, Cao F, Xiong Y, Zhou H. SP1 transcriptionally activates NLRP6 inflammasome and induces immune evasion and radioresistance in glioma cells. Int Immunopharmacol 2021; 98:107858. [PMID: 34147913 DOI: 10.1016/j.intimp.2021.107858] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 05/14/2021] [Accepted: 06/03/2021] [Indexed: 12/15/2022]
Abstract
Glioma accounts for approximately 80% of all malignant brain tumors. This study aimed to investigate the interaction between specificity protein 1 (SP1) and NLR family pyrin domain containing 6 (NLRP6) and their roles in the activity of glioma cells. Differentially expressed genes in glioma were identified using transcriptome analysis tools, and a protein-protein-interaction network was performed based on the DEGs. SP1 and NLRP6 were abundantly expressed in glioma cells and indicated unfavorable prognosis of patients according to the GEO datasets. SP1could bind to the promoter of NLRP6 and induce its transcriptional activity. Downregulation of SP1 reduced proliferation, migration and invasion of glioma U87 cells in vitro as well as tumorigenesis in vivo. The malignancy of cells was restored after NLRP6 upregulation. Downregulation of SP1 in glioma cells also increased proliferation of CD8+ T cells and the immune activity in U87 cells, and it reduced the radioresistance of U87 cells. However, the immune evasion and radioresistance of glioma cells were restored upon NLRP6 upregulation. NLRP6 mediated the innate immune pathway through an ASC/caspase-1/IL-1β axis. To conclude, this study suggested that SP1 interacts with NLRP6 inflammasome to enhance malignant behaviors, immune evasion and radioresistance in glioma cells.
Collapse
Affiliation(s)
- Yunhu Yu
- Clinical Research Center for Neurological Disease, People's Hospital of Honghuagang District of Zunyi, Zunyi 563000, Guizhou, PR China.
| | - Fang Cao
- Department of Cerebrovascular Disease, The First Affiliated Hospital of Zunyi Medical College, Zunyi 563000, Guizhou, PR China
| | - Yanquan Xiong
- Clinical Research Center for Neurological Disease, People's Hospital of Honghuagang District of Zunyi, Zunyi 563000, Guizhou, PR China
| | - Hang Zhou
- Clinical Research Center for Neurological Disease, People's Hospital of Honghuagang District of Zunyi, Zunyi 563000, Guizhou, PR China
| |
Collapse
|
15
|
Tavakoli F, Khatami SS, Momeni F, Azadbakht J, Ghasemi F. Cervical Cancer Diagnosis: Insights into Biochemical Biomarkers and Imaging Techniques. Comb Chem High Throughput Screen 2021; 24:605-623. [PMID: 32875976 DOI: 10.2174/1386207323666200901101955] [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: 03/31/2020] [Revised: 07/14/2020] [Accepted: 08/06/2020] [Indexed: 11/22/2022]
Abstract
Cervical malignancy is known as one of the important cancers which is originated from cervix. This malignancy has been observed in women infected with papillomavirus who had regular oral contraceptives, multiple pregnancies, and sexual relations. Early and fast cervical cancer diagnosis is known as two important aspects of cervical cancer therapy. Several investigations indicated that early and fast detection of cervical cancer could be associated with better treatment process and increasing survival rate of patients with this malignancy. Imaging techniques are very important diagnosis tools that could be employed for diagnosis and following responses to therapy in various cervical cancer stages. Multiple lines of evidence indicated that utilization of imaging techniques is related to some limitations (i.e. high cost, and invasive effects). Hence, it seems that along with using imaging techniques, finding and developing new biomarkers could be useful in the diagnosis and treatment of subjects with cervical cancer. Taken together, many studies showed that a variety of biomarkers including, several proteins, mRNAs, microRNAs, exosomes and polymorphisms might be introduced as prognostic, diagnostic and therapeutic biomarkers in cervical cancer therapy. In this review article, we highlighted imaging techniques as well as novel biomarkers for the diagnosis of cervical cancer.
Collapse
Affiliation(s)
- Fatemeh Tavakoli
- Department of Biotechnology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sara Sadat Khatami
- Department of Biotechnology, Faculty of Medicine, Arak University of Medical Sciences, Arak, Iran
| | - Fatemeh Momeni
- Isfahan Research Committee of Multiple Sclerosis, Alzahra Research Institute, Alzahra Hospital, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Javid Azadbakht
- Department of Radiology and Imaging, Kashan University of Medical Science, Kashan, Iran
| | - Faezeh Ghasemi
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran
| |
Collapse
|
16
|
Abstract
Extracellular acidification is a well-known driver of tumorigenesis that has been extensively studied. In contrast, the role of endosomal pH is novel and relatively unexplored. There is emerging evidence from a growing number of studies showing that the pH of endosomal compartments controls proliferation, migration, stemness, and sensitivity to chemoradiation therapy in a variety of tumors. Endosomes are a crucial hub, mediating cellular communication with the external environment. By finely regulating the sorting and trafficking of vesicular cargo for degradation or recycling, endosomal pH determines the fate of plasma membrane proteins, lipids, and extracellular signals including growth factor receptors and their ligands. Several critical regulators of endosomal pH have been identified, including multiple isoforms of the family of electroneutral Na+/H+ exchangers (NHE) such as NHE6 and NHE9. Recent studies have shed light on molecular mechanisms linking endosomal pH to cancer malignancy. Manipulating endosomal pH by epigenetic reprogramming, small molecules, or nanoparticles may offer promising new options in cancer therapy. In this review, we summarize evidence linking endosomal pH to cancer, with a focus on the role of endosomal Na+/H+ exchangers and how they affect the prognosis of cancer patients, and also suggest how regulation of endosomal pH may be exploited to develop new cancer therapies.
Collapse
|
17
|
Emami Nejad A, Najafgholian S, Rostami A, Sistani A, Shojaeifar S, Esparvarinha M, Nedaeinia R, Haghjooy Javanmard S, Taherian M, Ahmadlou M, Salehi R, Sadeghi B, Manian M. The role of hypoxia in the tumor microenvironment and development of cancer stem cell: a novel approach to developing treatment. Cancer Cell Int 2021; 21:62. [PMID: 33472628 PMCID: PMC7816485 DOI: 10.1186/s12935-020-01719-5] [Citation(s) in RCA: 372] [Impact Index Per Article: 93.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Revised: 12/11/2020] [Accepted: 12/16/2020] [Indexed: 12/13/2022] Open
Abstract
Hypoxia is a common feature of solid tumors, and develops because of the rapid growth of the tumor that outstrips the oxygen supply, and impaired blood flow due to the formation of abnormal blood vessels supplying the tumor. It has been reported that tumor hypoxia can: activate angiogenesis, thereby enhancing invasiveness and risk of metastasis; increase survival of tumor, as well as suppress anti-tumor immunity and hamper the therapeutic response. Hypoxia mediates these effects by several potential mechanisms: altering gene expression, the activation of oncogenes, inactivation of suppressor genes, reducing genomic stability and clonal selection. We have reviewed the effects of hypoxia on tumor biology and the possible strategiesto manage the hypoxic tumor microenvironment (TME), highlighting the potential use of cancer stem cells in tumor treatment.
Collapse
Affiliation(s)
- Asieh Emami Nejad
- Department of Biology, Payame Noor University (PNU), P.O.Box 19395-3697, Tehran, Iran
| | - Simin Najafgholian
- Department of Emergency Medicine, School of Medicine , Arak University of Medical Sciences, Arak, Iran
| | - Alireza Rostami
- Department of Surgery, School of Medicine Amiralmomenin Hospital, Arak University of Medical Sciences, Arak, Iran
| | - Alireza Sistani
- Department of Emergency Medicine, School of Medicine Valiasr Hospital, Arak University of Medical Sciences, Arak, Iran
| | - Samaneh Shojaeifar
- Department of Midwifery, Faculty of Nursing and Midwifery , Arak University of Medical Sciences , Arak, Iran
| | - Mojgan Esparvarinha
- Department of Immunology, School of Medicine , Tabriz University of Medical Sciences , Tabriz, Iran
| | - Reza Nedaeinia
- Pediatric Inherited Diseases Research Center, Research Institute for Primordial Prevention of Non-Communicable Disease , Isfahan University of Medical Sciences , Isfahan, Iran
| | - Shaghayegh Haghjooy Javanmard
- Applied Physiology Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences , Isfahan, Iran
| | - Marjan Taherian
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mojtaba Ahmadlou
- Sciences Medical of University Arak, Hospital Amiralmomenin, Center Development Research Clinical, Arak, Iran
| | - Rasoul Salehi
- Pediatric Inherited Diseases Research Center, Research Institute for Primordial Prevention of Non-Communicable Disease , Isfahan University of Medical Sciences , Isfahan, Iran.,Department of Genetics and Molecular Biology, School of Medicine , Isfahan University of Medical Sciences , Isfahan, Iran
| | - Bahman Sadeghi
- Department of Health and Community Medicine, School of Medicine, Arak University of Medical Sciences, Arak, 3848176341, Iran.
| | - Mostafa Manian
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran. .,Department of Medical Laboratory Science, Faculty of Medical Science Kermanshah Branch, Islamic Azad University, Imam Khomeini Campus, Farhikhtegan Bld., Shahid J'afari St., Kermanshah, 3848176341, Iran.
| |
Collapse
|
18
|
Li X, Wang H. Long Non-Coding RNA GABPB1-AS1 Augments Malignancy of Glioma Cells by Sequestering MicroRNA-330 and Reinforcing the ZNF367/Cell Cycle Signaling Pathway. Neuropsychiatr Dis Treat 2021; 17:2073-2087. [PMID: 34276213 PMCID: PMC8280909 DOI: 10.2147/ndt.s305182] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Accepted: 05/25/2021] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Deregulation of long non-coding RNAs (lncRNAs) is frequently relevant to the malignant phenotypical changes. This study aimed to explore the role of lncRNA GABPB1-AS1 in the malignancy of glioma cells. METHODS Abnormally expressed genes in glioma were analyzed using a GEO GSE2223 dataset. Short hairpin (sh) RNA silencing of GABPB1-AS1 was introduced in glioma cells to explore its correlation with the proliferation, apoptosis, and invasiveness of cancer cells. The target transcripts of GABPB1-AS1 were predicted by bioinformatics analyses. MicroRNA (miR)-330 inhibition was additionally introduced in the glioma cells after GABPB1-AS1 knockdown for rescue experiments. Animal studies were performed by inducing xenograft tumors in nude mice. RESULTS GABPB1-AS1 was highly expressed in the glioma tissues and associated with advanced WHO grades. GABPB1-AS1 knockdown reduced proliferation and invasiveness of glioma cells in vitro and in vivo. miR-330 was a target transcript of GABPB1-AS1. miR-330 inhibition restored the malignancy of glioma cells. miR-330 directly bound to ZNF367. ZNF367 was highly expressed in glioma tissues and positively correlated with GABPB1-AS1 expression, and it was relevant to the cell cycle signaling pathway. Downregulation of GABPB1-AS1 reduced the expression of ZNF367 and reduced the levels of cell cycle-related proteins PCNA, CDC20, CDC7 and CCNA1 in cells. CONCLUSION This study demonstrated that GABPB1-AS1 competitively bound to miR-330 and de-repressed ZNF367 expression, leading to activation of the cell cycle signaling pathway and the growth and metastasis of glioma cells.
Collapse
Affiliation(s)
- Xiulong Li
- Department of Neurosurgery, Caoxian People's Hospital, Heze, 274400, Shandong, People's Republic of China
| | - Hongfeng Wang
- Department of Neurosurgery, Yantai Harbour Hospital, Yantai, 264000, Shandong, People's Republic of China
| |
Collapse
|
19
|
Abstract
Purpose of review This review focuses on the development and progression of glioblastoma through the brain and glioma microenvironment. Specifically we highlight how the tumor microenvironment contributes to the hallmarks of cancer in hopes of offering novel therapeutic options and tools to target this microenvironment. Recent findings The hallmarks of cancer, which represent elements of cancers that contribute to the disease's malignancy, yet elements within the brain tumor microenvironment, such as other cellular types as well as biochemical and biophysical cues that can each uniquely affect tumor cells, have not been well-described in this context and serve as potential targets for modulation. Summary Here, we highlight how the brain tumor microenvironment contributes to the progression and therapeutic response of tumor cells. Specifically, we examine these contributions through the lens of Hanahan & Weinberg's Hallmarks of Cancer in order to identify potential novel targets within the brain that may offer a means to treat brain cancers, including the deadliest brain cancer, glioblastoma.
Collapse
|
20
|
LingHu HR, Luo H, Gang L. Bufalin Induces Glioma Cell Death by Apoptosis or Necroptosis. Onco Targets Ther 2020; 13:4767-4778. [PMID: 32581545 PMCID: PMC7274536 DOI: 10.2147/ott.s242567] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 05/04/2020] [Indexed: 11/23/2022] Open
Abstract
Introduction Bufalin is a component of Chinese traditional medicine, Chansu, which is reported to induce cell death among various kinds of tumors. Apoptosis evasion is a common problem of cancer treatment. Materials and Methods The proliferation of U-87 and U-373 treated by bufalin combined with or without apoptosis inhibitor was detected by MTT assay. The protein levels related to apoptosis and necroptosis were measured by Western blotting. Immunoprecipitation (IP) was applied for monitoring the formation of necrosome. The gene knockdown by CRISPR/Cas9 was applied to determine the roles of the proteins in apoptosis and necroptosis. Results In this study, we found that bufalin could induce apoptosis or necroptosis when U-87 and U-373 escaped from apoptosis. Bufalin triggered cell death by upregulating tumor necrosis factor (TNF) -α, TNF receptor 1 (TNFR1) and receptor-interacting protein 1 (RIPK1). Antagonizing cellular inhibitor of apoptosis 1 (cIAP1) and cIAP2 were also contributory. Caspase-8 activation led to apoptosis. When caspase-8 was functionally lost, necrosome consisted of RIPK1, receptor-interacting protein 3 (RIPK3) and mixed lineage kinase domain-like protein (MLKL) formed and necroptosis happened. The knockdown of above genes or the drug treatment confirmed the mechanism of bufalin-induced cell death. Cytotoxicity of bufalin to caspase-8 knockdown cell lines made control cell lines more sensitive to bufalin in their mixture. Discussion The cytotoxicity of bufalin to U-87 and U-373 was by inducing apoptosis or necroptosis when they were sensitive to apoptosis or not. The results indicated that seeking for treatments that could induce apoptosis and necroptosis was a good solution for the tumor evasion of apoptosis.
Collapse
Affiliation(s)
- Hai Rui LingHu
- Department of Neurosurgery and Neurocritical Care, Beijing Chaoyang Integrative Medicine Emergency Medical Center, Beijing 100022, People's Republic of China
| | - Hui Luo
- Characteristic Medical Center of Chinese People's Armed Police Force (PAP), Tianjin 300162, People's Republic of China
| | - Lin Gang
- Characteristic Medical Center of Chinese People's Armed Police Force (PAP), Tianjin 300162, People's Republic of China
| |
Collapse
|
21
|
Ashrafizadeh M, Najafi M, Makvandi P, Zarrabi A, Farkhondeh T, Samarghandian S. Versatile role of curcumin and its derivatives in lung cancer therapy. J Cell Physiol 2020; 235:9241-9268. [PMID: 32519340 DOI: 10.1002/jcp.29819] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 04/24/2020] [Accepted: 05/12/2020] [Indexed: 12/24/2022]
Abstract
Lung cancer is a main cause of death all over the world with a high incidence rate. Metastasis into neighboring and distant tissues as well as resistance of cancer cells to chemotherapy demand novel strategies in lung cancer therapy. Curcumin is a naturally occurring nutraceutical compound derived from Curcuma longa (turmeric) that has great pharmacological effects, such as anti-inflammatory, neuroprotective, and antidiabetic. The excellent antitumor activity of curcumin has led to its extensive application in the treatment of various cancers. In the present review, we describe the antitumor activity of curcumin against lung cancer. Curcumin affects different molecular pathways such as vascular endothelial growth factors, nuclear factor-κB (NF-κB), mammalian target of rapamycin, PI3/Akt, microRNAs, and long noncoding RNAs in treatment of lung cancer. Curcumin also can induce autophagy, apoptosis, and cell cycle arrest to reduce the viability and proliferation of lung cancer cells. Notably, curcumin supplementation sensitizes cancer cells to chemotherapy and enhances chemotherapy-mediated apoptosis. Curcumin can elevate the efficacy of radiotherapy in lung cancer therapy by targeting various signaling pathways, such as epidermal growth factor receptor and NF-κB. Curcumin-loaded nanocarriers enhance the bioavailability, cellular uptake, and antitumor activity of curcumin. The aforementioned effects are comprehensively discussed in the current review to further direct studies for applying curcumin in lung cancer therapy.
Collapse
Affiliation(s)
- Milad Ashrafizadeh
- Department of Basic Science, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran
| | - Masoud Najafi
- Radiology and Nuclear Medicine Department, School of Paramedical Sciences, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Pooyan Makvandi
- Institute for Polymers, Composites and Biomaterials (IPCB), National Research Council (CNR), Naples, Italy
| | - Ali Zarrabi
- Sabanci University Nanotechnology Research and Application Center (SUNUM), Tuzla, Istanbul, Turkey
| | - Tahereh Farkhondeh
- Cardiovascular Diseases Research Center, Birjand University of Medical Sciences, Birjand, Iran
| | - Saeed Samarghandian
- Healthy Ageing Research Center, Neyshabur University of Medical Sciences, Neyshabur, Iran
| |
Collapse
|
22
|
Harguindey S, Polo Orozco J, Alfarouk KO, Devesa J. Hydrogen Ion Dynamics of Cancer and a New Molecular, Biochemical and Metabolic Approach to the Etiopathogenesis and Treatment of Brain Malignancies. Int J Mol Sci 2019; 20:ijms20174278. [PMID: 31480530 PMCID: PMC6747469 DOI: 10.3390/ijms20174278] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 08/28/2019] [Accepted: 08/30/2019] [Indexed: 12/13/2022] Open
Abstract
The treatment of cancer has been slowly but steadily progressing during the last fifty years. Some tumors with a high mortality in the past are curable nowadays. However, there is one striking exception: glioblastoma multiforme. No real breakthrough has been hitherto achieved with this tumor with ominous prognosis and very short survival. Glioblastomas, being highly glycolytic malignancies are strongly pH-dependent and driven by the sodium hydrogen exchanger 1 (NHE1) and other proton (H+) transporters. Therefore, this is one of those pathologies where the lessons recently learnt from the new pH-centered anticancer paradigm may soon bring a promising change to treatment. This contribution will discuss how the pH-centric molecular, biochemical and metabolic perspective may introduce some urgently needed and integral novel treatments. Such a prospective therapeutic approach for malignant brain tumors is developed here, either to be used alone or in combination with more standard therapies.
Collapse
Affiliation(s)
| | | | - Khalid O Alfarouk
- Al-Ghad International Colleges for Applied Medical Sciences, Al-Madinah Al-Munawarah 42316, Saudi Arabia
- Alfarouk Biomedical Research LLC, Tampa, FL 33617, USA
| | - Jesús Devesa
- Scientific Direction, Foltra Medical Centre, 15886 Teo, Spain
| |
Collapse
|