1
|
Zhou Y, Qin L, Li C, Zhu D, Liu B. EGCG enhances antitumor effect of apatinib in nonsmall cell lung cancer by targeting VEGF signaling to inhibit glycolysis. Drug Dev Res 2024; 85:e22239. [PMID: 39397333 DOI: 10.1002/ddr.22239] [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: 03/03/2024] [Revised: 06/19/2024] [Accepted: 07/15/2024] [Indexed: 10/15/2024]
Abstract
Nonsmall cell lung cancer (NSCLC), one of the most aggressive malignancies globally, is characterized by poor prognosis and limited life expectancy. Epigallocatechin-3-gallate (EGCG), a natural polyphenol found in green tea, has emerged as a promising anticancer agent due to its potent antitumor properties. However, the role and the underlying mechanisms of EGCG in NSCLC remain poorly understood. Hence, this research aimed to explore the effect of EGCG on the antitumor effect of apatinib in NSCLC through vascular endothelial growth factor (VEGF)-regulated glycolysis. Cell Counting Kit-8 (CCK-8), 5-ethynyl-2'-deoxyuridine staining, wound healing, transwell, terminal deoxynucleotidyl transferase dUTP nick-end labeling, and flow cytometry assays were carried out to evaluate the proliferation, migration, invasion, and apoptosis of H1299 cells, respectively. Furthermore, western blot analysis was used to detect the expressions of VEGF, p-vascular endothelial growth factor receptor-2, hypoxia-inducible factor 1α, neuropilin-1, phosphorylated-phosphatidylinositol 3-kinase, and phosphorylated-AKT. The transfection efficiency of H1299 cells with VEGF overexpression plasmid was also assessed by western blot analysis. Glycolysis was analyzed by estimating extracellular acidification rate, lactate concentration, glucose uptake, and the expressions of lactate dehydrogenase A, pyruvate kinase M2, and hexokinase 2. The results demonstrated that VEGF activated glycolysis in NSCLC cells. EGCG alone and apatinib alone or in combination inhibited cell viability, proliferation, invasion, migration, and glycolysis whereas promoted apoptosis in NSCLC cells. EGCG regulated glycolysis levels in NSCLC through VEGF overexpression, and enhanced the antitumor effect of apatinib in NSCLC through VEGF-regulated glycolysis. Taken together, EGCG strengthened the protective effects of apatinib in NSCLC through glycolysis mediated by VEGF.
Collapse
Affiliation(s)
- Yue Zhou
- College of Pharmacy, Guizhou University, Guiyang, China
- Guizhou Engineering Laboratory for Synthetic Drugs, Guizhou University, Guiyang, China
| | - Liqing Qin
- College of Pharmacy, Guizhou University, Guiyang, China
| | - Chengpeng Li
- College of Pharmacy, Guizhou University, Guiyang, China
- Guizhou Engineering Laboratory for Synthetic Drugs, Guizhou University, Guiyang, China
| | - Danxue Zhu
- College of Pharmacy, Guizhou University, Guiyang, China
| | - Bo Liu
- College of Pharmacy, Guizhou University, Guiyang, China
| |
Collapse
|
2
|
Hou S, Chen Y, Jin C, Lin N. Integrative analysis of bulk RNA-seq and scRNA-seq data indicates the prognostic and immunologic values of SERPINH1 in glioma. ENVIRONMENTAL TOXICOLOGY 2024; 39:3654-3665. [PMID: 38506564 DOI: 10.1002/tox.24192] [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: 10/10/2023] [Revised: 01/29/2024] [Accepted: 02/25/2024] [Indexed: 03/21/2024]
Abstract
BACKGROUND SERPINH1 is abnormally expressed in multiple cancers and is associated with malignant progression. However, few reports detail its role in the etiopathogenesis of glioma. Hence, the aim of this article was to investigate the potential value of SERPINH1 in glioma using an integrative analysis. METHODS Data of RNA-seq and scRNA-seq was obtained and evaluated using online databases. The expression of SERPINH1 was confirmed by qRT-PCR and immunohistochemistry. The prognostic value of SERPINH1 was evaluated using univariate and multivariate Cox regression analyses. SERPINH1-related signaling pathways and the interaction of SERPINH1 with immunity were also investigated. RESULTS SERPINH1 exhibited a markedly elevated expression in glioma compared to normal brain tissues in the online databases. Similar results were confirmed by qRT-PCR and immunohistochemistry. SERPINH1 was found to be an independent prognosis factor, and high expression of SERPINH1 indicated poor survival. Moreover, a nomogram was constructed to predict prognosis more accurately and intuitively. GSEA analysis showed that SERPINH1 was involved in seven signaling pathways, including JAK-STAT pathway. Further analysis indicated SERPINH1 was significantly associated with immunity, especially in low-grade glioma. Additionally, an examination of scRNA-seq data revealed that SERPINH1 was primarily expressed in T cells of the CD4+ and CD8+ subsets. CONCLUSIONS SERPINH1 is a key biomarker of glioma prognosis and is immunologically relevant, which provides additional options for targeted therapy of glioma.
Collapse
Affiliation(s)
- Shiqiang Hou
- Department of Neurosurgery, The Affiliated Chuzhou Hospital of Anhui Medical University, The First People's Hospital of Chuzhou, Chuzhou, China
| | - Yinan Chen
- Department of Neurosurgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Chunjing Jin
- Laboratory Medicine Center, The Affiliated Chuzhou Hospital of Anhui Medical University, The First People's Hospital of Chuzhou, Chuzhou, China
| | - Ning Lin
- Department of Neurosurgery, The Affiliated Chuzhou Hospital of Anhui Medical University, The First People's Hospital of Chuzhou, Chuzhou, China
| |
Collapse
|
3
|
Su MC, Lee AM, Zhang W, Maeser D, Gruener RF, Deng Y, Huang RS. Computational Modeling to Identify Drugs Targeting Metastatic Castration-Resistant Prostate Cancer Characterized by Heightened Glycolysis. Pharmaceuticals (Basel) 2024; 17:569. [PMID: 38794139 PMCID: PMC11124089 DOI: 10.3390/ph17050569] [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: 03/29/2024] [Revised: 04/22/2024] [Accepted: 04/26/2024] [Indexed: 05/26/2024] Open
Abstract
Metastatic castration-resistant prostate cancer (mCRPC) remains a deadly disease due to a lack of efficacious treatments. The reprogramming of cancer metabolism toward elevated glycolysis is a hallmark of mCRPC. Our goal is to identify therapeutics specifically associated with high glycolysis. Here, we established a computational framework to identify new pharmacological agents for mCRPC with heightened glycolysis activity under a tumor microenvironment, followed by in vitro validation. First, using our established computational tool, OncoPredict, we imputed the likelihood of drug responses to approximately 1900 agents in each mCRPC tumor from two large clinical patient cohorts. We selected drugs with predicted sensitivity highly correlated with glycolysis scores. In total, 77 drugs predicted to be more sensitive in high glycolysis mCRPC tumors were identified. These drugs represent diverse mechanisms of action. Three of the candidates, ivermectin, CNF2024, and P276-00, were selected for subsequent vitro validation based on the highest measured drug responses associated with glycolysis/OXPHOS in pan-cancer cell lines. By decreasing the input glucose level in culture media to mimic the mCRPC tumor microenvironments, we induced a high-glycolysis condition in PC3 cells and validated the projected higher sensitivity of all three drugs under this condition (p < 0.0001 for all drugs). For biomarker discovery, ivermectin and P276-00 were predicted to be more sensitive to mCRPC tumors with low androgen receptor activities and high glycolysis activities (AR(low)Gly(high)). In addition, we integrated a protein-protein interaction network and topological methods to identify biomarkers for these drug candidates. EEF1B2 and CCNA2 were identified as key biomarkers for ivermectin and CNF2024, respectively, through multiple independent biomarker nomination pipelines. In conclusion, this study offers new efficacious therapeutics beyond traditional androgen-deprivation therapies by precisely targeting mCRPC with high glycolysis.
Collapse
Affiliation(s)
- Mei-Chi Su
- Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455, USA; (M.-C.S.); (A.M.L.); (R.F.G.)
| | - Adam M. Lee
- Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455, USA; (M.-C.S.); (A.M.L.); (R.F.G.)
| | - Weijie Zhang
- Bioinformatics and Computational Biology, University of Minnesota, Minneapolis, MN 55455, USA; (W.Z.); (D.M.)
| | - Danielle Maeser
- Bioinformatics and Computational Biology, University of Minnesota, Minneapolis, MN 55455, USA; (W.Z.); (D.M.)
| | - Robert F. Gruener
- Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455, USA; (M.-C.S.); (A.M.L.); (R.F.G.)
| | - Yibin Deng
- Department of Urology, Masonic Cancer Center, University of Minnesota Medical School, Minneapolis, MN 55455, USA;
| | - R. Stephanie Huang
- Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455, USA; (M.-C.S.); (A.M.L.); (R.F.G.)
- Bioinformatics and Computational Biology, University of Minnesota, Minneapolis, MN 55455, USA; (W.Z.); (D.M.)
| |
Collapse
|
4
|
Kaur B, Blavo C, Parmar MS. Ivermectin: A Multifaceted Drug With a Potential Beyond Anti-parasitic Therapy. Cureus 2024; 16:e56025. [PMID: 38606261 PMCID: PMC11008553 DOI: 10.7759/cureus.56025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Accepted: 03/11/2024] [Indexed: 04/13/2024] Open
Abstract
Ivermectin was first discovered in the 1970s by Japanese microbiologist Satoshi Omura and Irish parasitologist William C. Campbell. Ivermectin has become a versatile pharmaceutical over the past 50 years. Ivermectin is a derivative of avermectin originally used to treat parasitic infections. Emerging literature has suggested that its role goes beyond this and may help treat inflammatory conditions, viral infections, and cancers. Ivermectin's anti-parasitic, anti-inflammatory, anti-viral, and anticancer effects were explored. Its traditional mechanism of action in parasitic diseases, such as scabies and malaria, rests on its ability to interfere with the glutamate-gated chloride channels in invertebrates and the lack of P-glycoprotein in many parasites. More recently, it has been discovered that the ability of ivermectin to block the nuclear factor kappa-light-chain enhancer of the activated B (NF-κB) pathway that modulates the expression and production of proinflammatory cytokines is implicated in its role as an anti-inflammatory agent to treat rosacea. Ivermectin has also been evaluated for treating infections caused by viruses, such as SARS-CoV-2 and adenoviruses, through inhibition of viral protein transportation and acting on the importin α/β1 interface. It has also been suggested that ivermectin can inhibit the proliferation of tumorigenic cells through various pathways that lead to the management of certain cancers. The review aimed to evaluate its multifaceted effects and potential clinical applications beyond its traditional use as an anthelmintic agent.
Collapse
Affiliation(s)
- Baneet Kaur
- Department of Medicine, Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Clearwater, USA
| | - Cyril Blavo
- Department of Public Health, Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Clearwater, USA
| | - Mayur S Parmar
- Department of Foundational Sciences, Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Clearwater, USA
| |
Collapse
|
5
|
Hu X, Ju Y, Zhang YK. Ivermectin as a potential therapeutic strategy for glioma. J Neurosci Res 2024; 102:e25254. [PMID: 37814994 DOI: 10.1002/jnr.25254] [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: 04/17/2023] [Revised: 09/22/2023] [Accepted: 09/24/2023] [Indexed: 10/11/2023]
Abstract
Ivermectin (IVM), a semi-synthetic macrolide parasiticide, has demonstrated considerable effectiveness in combating internal and external parasites, particularly nematodes and arthropods. Its remarkable ability to control parasites has earned it significant recognition, culminating in Satoshi Omura and William C. Campbell's receipt of the 2015 Nobel Prize in Physiology or Medicine for their contributions to the development of IVM. In recent years, investigations have revealed that IVM possesses antitumor properties. It can suppress the growth of various cancer cells, including glioma, through a multitude of mechanisms such as selective targeting of tumor-specific proteins, inducing programmed cell death, and modulation of tumor-related signaling pathways. Hence, IVM holds tremendous potential as a novel anticancer drug. This review seeks to provide an overview of the underlying mechanisms that enable IVM's capacity to suppress glioma. Furthermore, it aims to elucidate the challenges and prospects associated with utilizing IVM as a new anticancer agent.
Collapse
Affiliation(s)
- Xing Hu
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, PR China
| | - Yan Ju
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, PR China
| | - Yue-Kang Zhang
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, PR China
| |
Collapse
|
6
|
Castillejos-López M, Torres-Espíndola LM, Huerta-Cruz JC, Flores-Soto E, Romero-Martinez BS, Velázquez-Cruz R, Higuera-Iglesias A, Camarena Á, Torres-Soria AK, Salinas-Lara C, Fernández-Plata R, Alvarado-Vásquez N, Solís-Chagoyán H, Ruiz V, Aquino-Gálvez A. Ivermectin: A Controversial Focal Point during the COVID-19 Pandemic. Life (Basel) 2022; 12:1384. [PMID: 36143420 PMCID: PMC9502658 DOI: 10.3390/life12091384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 08/25/2022] [Accepted: 08/31/2022] [Indexed: 01/08/2023] Open
Abstract
The SARS-CoV-2 pandemic has confirmed the apocalyptic predictions that virologists have been making for several decades. The challenge the world is facing is that of trying to find a possible treatment, and a viable and expedient option for addressing this challenge is the repurposing of drugs. However, in some cases, although these drugs are approved for use in humans, the mechanisms of action involved are unknown. In this sense, to justify its therapeutic application to a new disease, it is ideal, but not necessary, to know the basic mechanisms of action involved in a drug's biological effects. This review compiled the available information regarding the various effects attributed to Ivermectin. The controversy over its use for the treatment of COVID-19 is demonstrated by this report that considers the proposal unfeasible because the therapeutic doses proposed to achieve this effect cannot be achieved. However, due to the urgent need to find a treatment, an exhaustive and impartial review is necessary in order to integrate the knowledge that exists, to date, of the possible mechanisms through which the treatment may be helpful in defining safe doses and schedules of Ivermectin.
Collapse
Affiliation(s)
- Manuel Castillejos-López
- Departamento de Epidemiología y Estadística, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City 14080, Mexico
| | | | - Juan Carlos Huerta-Cruz
- Unidad de Investigación en Farmacología, Instituto Nacional de Enfermedades Respiratorias Ismael Cosio Villegas, Mexico City 14080, Mexico
| | - Edgar Flores-Soto
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
| | - Bianca S. Romero-Martinez
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
| | - Rafael Velázquez-Cruz
- Laboratorio de Genómica del Metabolismo Óseo, Instituto Nacional de Medicina Genómica, Mexico City 14610, Mexico
| | - Anjarath Higuera-Iglesias
- Departamento de Investigación en Epidemiología Clínica, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City 14080, Mexico
| | - Ángel Camarena
- Laboratorio de HLA, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City 14080, Mexico
| | - Ana Karen Torres-Soria
- Red MEDICI, Carrera de Médico Cirujano, Facultad de Estudios Superiores de Iztacala UNAM, Mexico City 54090, Mexico
| | - Citlaltepetl Salinas-Lara
- Red MEDICI, Carrera de Médico Cirujano, Facultad de Estudios Superiores de Iztacala UNAM, Mexico City 54090, Mexico
| | - Rosario Fernández-Plata
- Departamento de Epidemiología y Estadística, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City 14080, Mexico
| | - Noé Alvarado-Vásquez
- Departamento de Bioquímica, Instituto Nacional de Enfermedades Respiratorias Ismael Cosio Villegas, Mexico City 14080, Mexico
| | - Héctor Solís-Chagoyán
- Subdirección de Investigaciones Clínicas, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Mexico City 14370, Mexico
| | - Víctor Ruiz
- Laboratorio de Biología Molecular, Departamento de Fibrosis Pulmonar, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City 14080, Mexico
| | - Arnoldo Aquino-Gálvez
- Laboratorio de Biología Molecular, Departamento de Fibrosis Pulmonar, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City 14080, Mexico
| |
Collapse
|
7
|
The In Vitro Effect of Psoralen on Glioma Based on Network Pharmacology and Potential Target Research. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:1952891. [PMID: 36065261 PMCID: PMC9440786 DOI: 10.1155/2022/1952891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 06/30/2022] [Accepted: 07/05/2022] [Indexed: 11/23/2022]
Abstract
Glioma is an aggressive tumor, currently there is no satisfactory management available. Psoralen, as a natural product, has been found to have an effect of treating cancer in recent years, but its effect on glioma has not been explored. In this study, we investigated the in vitro inhibition effect and potential targets of psoralen on glioma through network pharmacology and in vitro glioma treatment experiments. First, we used network pharmacology to preliminarily predict the 21 core genes of psoralen in the treatment of glioma, including PIK3CA, PIK3CB, PIK3CG, and JAK2. The CCK-8 method was used to detect the effect of psoralen on the proliferation of glioma U87 and U251 cells, and the results showed that psoralen could significantly inhibit the proliferation of U87 and U251 cells. The flow cytometry was used to detect the apoptosis and cell cycle changes, and it was found that psoralen could significantly promote the early apoptosis of U87 and U251 cells and had a significant cycle arrest effect on the two cells. The cell scratch test showed that psoralen could significantly inhibit the migration of U87 and U251 cells. The relative expression levels of PIK3CA, PIK3CB, PIK3CG, and JAK2 were analyzed by Real-time Quantitative polymerase chain reaction (QT-PCR), and the results showed that psoralen could inhibit the gene expression of PIK3CA, PIK3CB, PIK3CG, and JAK2. Later, Western blotting (WB) experiments showed that psoralen could inhibit the protein expressions of PI3K and JAK2. This study has preliminarily explored and verified the antiglioma effect of psoralen in the form of inhibiting cell proliferation and migration, promoting cell apoptosis and organizing cell cycle in vitro. And may play a role by inhibiting the expression of PIK3CA, PIK3CB, PIK3CG, JAK2 gene and PI3K, JAK2 protein, psoralen has become a potential antiglioma drug.
Collapse
|