1
|
Verma R, Zeyaullah M, Singh V, Saxena PS, Koch B, Kumar M. Chitosan-Functionalized Fluorescent Calcium Carbonate Nanoparticle Loaded with Methotrexate: Future Theranostics for Triple Negative Breast Cancer. ACS Biomater Sci Eng 2025; 11:981-999. [PMID: 39883480 DOI: 10.1021/acsbiomaterials.4c02390] [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] [Indexed: 01/31/2025]
Abstract
Herein, fluorescent calcium carbonate nanoclusters encapsulated with methotrexate (Mtx) and surface functionalized with chitosan (25 nm) (@Calmat) have been developed for the imaging and treatment of triple-negative breast cancer (TNBC). These biocompatible, pH-sensitive nanoparticles demonstrate significant potential for targeted therapy and diagnostic applications. The efficacy of nanoparticles (NPs) was evaluated in MDA-MB-231 TNBC cell lines. The enhanced permeability and retention effect facilitated the accumulation of NPs, in tumor-bearing rats, as confirmed by in vivo fluorescence imaging. Treatment with @Calmat resulted in a marked reduction in pro-inflammatory cytokines, with levels of IL-6 (1225 ± 67 pg/mL), IL-1β (379 ± 69 pg/mL), and TNF-α (14.1 ± 2 pg/mL), in contrast to the diseased control group (IL-6: 2223 ± 99; IL-1β: 1632 ± 90; TNF-α: 40 ± 3 pg/mL). A similar trend was observed for liver and kidney function biomarkers. Mechanistic studies revealed that @Calmat treatment activates the Bax/Bcl-2 signaling pathway, leading to cell cycle arrest in the G1 phase and subsequent late-phase apoptosis. As a result, the tumor inhibition rate reached 88%, with 80% of treated rats surviving beyond 100 days. These findings highlight the strong potential of @Calmat as a dual-function theranostic agent for the management of TNBC.
Collapse
Affiliation(s)
- Rinki Verma
- School of Biomedical Engineering, IIT (BHU), Varanasi 221005, India
| | - Md Zeyaullah
- Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi 221005, India
| | - Virendra Singh
- Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi 221005, India
| | - Preeti Suman Saxena
- Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi 221005, India
| | - Biplob Koch
- Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi 221005, India
| | - Manoj Kumar
- Nano 2 Micro Material Design Lab, Department of Chemical Engineering and Technology, IIT (BHU), Varanasi 221005, India
| |
Collapse
|
2
|
Klaimanee E, Temram T, Ratanaphan A, Saithong S, Sooksawat D, Samphao A, Yakiyama Y, Sakurai H, Konno T, Tantirungrotechai Y, Choojun K, Leesakul N. Iridium(III) coordination compounds based on organophosphorus ancillary ligands showing cytotoxicity against breast cancer cells and Fe(III) luminescent sensing. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2025; 325:125150. [PMID: 39305800 DOI: 10.1016/j.saa.2024.125150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 09/11/2024] [Accepted: 09/13/2024] [Indexed: 11/10/2024]
Abstract
Three phosphorescent iridium(III) complexes consisting bis-diphosphine ligands were prepared and characterized by single-crystal XRD, CHN analysis, spectroscopic techniques, cyclic voltammetry, and DFT. The synthesized complexes were the three monomeric [Ir(ppy)2(L1)Cl] (1), [Ir(ppy)2(L2)]Cl (2) and [Ir(ppy)2(L3)]Cl (3) where L1 = bis-(diphenylphosphino)methane (dppm), L2 = bis-(diphenylphosphino)propane (dppp) and L3 = bis-(diphenylphosphino)benzene (dppbe). Complexes 1-3 gave an absorption band between 240 to 380 nm in both CH2Cl2 and DMSO, which is assigned as a charge transfer transition based on theoretical calculation. They showed a blue-green emission at 460-520 nm in DMSO with an absolute quantum efficiency of 0.013-0.046 at room temperature. The selective photo-induced electron transfer (PET) by Fe3+ in DMSO, was studied to obey the Rehm-Weller principle. The 1:1 binding soichiometry between 1-3 and Fe3+ was established by Job's plot. The binding constants (Ka) were determined using the Benesi-Hildebrand plot. All the complexes are extremely more potent than cisplatin for in vitro antiproliferative activity towards the human breast cancer cells, HCC1937, MCF-7, and MDA-MB-231. The values of IC50 were in the range of 0.077-0.485 μM, and 1 exhibited the most effective IC50 against MDA-MB-231 cell line, the triple-negative breast cancer cell. Their lipophilicities (log P) were also examined to explain the penetration ability of the studied complexes towards cell barriers, and transport to the molecular target.
Collapse
Affiliation(s)
- Ekkapong Klaimanee
- Division of Physical Science and Center of Excellence for Innovation in Chemistry, Faculty of Science, Prince of Songkla University, Hat-Yai, Songkhla 90112, Thailand
| | - Thitirat Temram
- Division of Physical Science and Center of Excellence for Innovation in Chemistry, Faculty of Science, Prince of Songkla University, Hat-Yai, Songkhla 90112, Thailand
| | - Adisorn Ratanaphan
- Department of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences, Prince of Songkla University, Hat Yai, Songkhla 90112, Thailand
| | - Saowanit Saithong
- Division of Physical Science and Center of Excellence for Innovation in Chemistry, Faculty of Science, Prince of Songkla University, Hat-Yai, Songkhla 90112, Thailand; Medical Science Research and Innovation Institute, Research and Development Office, Prince of Songkla University, Hat-Yai 90112, Thailand
| | - Dhassida Sooksawat
- Division of Physical Science and Center of Excellence for Innovation in Chemistry, Faculty of Science, Prince of Songkla University, Hat-Yai, Songkhla 90112, Thailand
| | - Anchalee Samphao
- Department of Chemistry, Faculty of Science, Ubonratchathani University, Ubonratchathani, 34190, Thailand
| | - Yumi Yakiyama
- Division of Applied Chemistry, Graduate School of Engineering, and Innovative Catalysis Science Division, Institute for Open and Transdisciplinary Research Initiatives (ICS-OTRI), Osaka University, Suita, Osaka 565-0871, Japan
| | - Hidehiro Sakurai
- Division of Applied Chemistry, Graduate School of Engineering, and Innovative Catalysis Science Division, Institute for Open and Transdisciplinary Research Initiatives (ICS-OTRI), Osaka University, Suita, Osaka 565-0871, Japan
| | - Takumi Konno
- Department of Chemistry, Graduate School of Science, Osaka University, Toyonaka, Osaka 560-0043, Japan; Department of Chemistry, College of Science, National Taiwan Normal University, Taipei 11677, Taiwan
| | - Yuthana Tantirungrotechai
- Thammasat University Research Unit in Innovation of Molecular Hybrid for Biomedical Application and Division of Chemistry, Faculty of Science and Technology, Thammasat University, Pathumthani 12120, Thailand
| | - Kittisak Choojun
- Catalytic Chemistry Research Unit, School of Science, King Mongkut's Institute of Technology Ladkrabang, Chalongkrung Road, Ladkrabang, Bangkok 10520, Thailand
| | - Nararak Leesakul
- Division of Physical Science and Center of Excellence for Innovation in Chemistry, Faculty of Science, Prince of Songkla University, Hat-Yai, Songkhla 90112, Thailand.
| |
Collapse
|
3
|
Xiong S, Song K, Xiang H, Luo G. Dual-target inhibitors based on ERα: Novel therapeutic approaches for endocrine resistant breast cancer. Eur J Med Chem 2024; 270:116393. [PMID: 38588626 DOI: 10.1016/j.ejmech.2024.116393] [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: 05/25/2023] [Revised: 04/04/2024] [Accepted: 04/04/2024] [Indexed: 04/10/2024]
Abstract
Estrogen receptor alpha (ERα), a nuclear transcription factor, is a well-validated therapeutic target for more than 70% of all breast cancers (BCs). Antagonizing ERα either by selective estrogen receptor modulators (SERMs) or selective estrogen receptor degraders (SERDs) forms the foundation of endocrine therapy and has achieved great success in the treatment of ERα positive (ERα+) BCs. Unfortunately, despite initial effectiveness, endocrine resistance eventually emerges in up to 30% of ERα+ BC patients and remains a significant medical challenge. Several mechanisms implicated in endocrine resistance have been extensively studied, including aberrantly activated growth factor receptors and downstream signaling pathways. Hence, the crosstalk between ERα and another oncogenic signaling has led to surge of interest to develop combination therapies and dual-target single agents. This review briefly introduces the synergisms between ERα and another anticancer target and summarizes the recent advances of ERα-based dual-targeting inhibitors from a medicinal chemistry perspective. Accordingly, their rational design strategies, structure-activity relationships (SARs) and biological activities are also dissected to provide some perspectives on future directions for ERα-based dual target drug discovery in BC therapy.
Collapse
Affiliation(s)
- Shuangshuang Xiong
- Jiangsu Key Laboratory of Drug Design and Optimization, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Ke Song
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Hua Xiang
- Jiangsu Key Laboratory of Drug Design and Optimization, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China.
| | - Guoshun Luo
- Jiangsu Key Laboratory of Drug Design and Optimization, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China.
| |
Collapse
|
4
|
Tripathi AK, Desai PP, Tyagi A, Lampe JB, Srivastava Y, Donkor M, Jones HP, Dzyuba SV, Crossley E, Williams NS, Vishwanatha JK. Short peptides based on the conserved regions of MIEN1 protein exhibit anticancer activity by targeting the MIEN1 signaling pathway. J Biol Chem 2024; 300:105680. [PMID: 38272230 PMCID: PMC10878790 DOI: 10.1016/j.jbc.2024.105680] [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: 09/23/2023] [Revised: 01/06/2024] [Accepted: 01/09/2024] [Indexed: 01/27/2024] Open
Abstract
Migration and invasion enhancer 1 (MIEN1) overexpression characterizes several cancers and facilitates cancer cell migration and invasion. Leveraging conserved immunoreceptor tyrosine-based activation motif and prenylation motifs within MIEN1, we identified potent anticancer peptides. Among them, bioactive peptides LA3IK and RP-7 induced pronounced transcriptomic and protein expression changes at sub-IC50 concentrations. The peptides effectively inhibited genes and proteins driving cancer cell migration, invasion, and epithelial-mesenchymal transition pathways, concurrently suppressing epidermal growth factor-induced nuclear factor kappa B nuclear translocation in metastatic breast cancer cells. Specifically, peptides targeted the same signal transduction pathway initiated by MIEN1. Molecular docking and CD spectra indicated the formation of MIEN1-peptide complexes. The third-positioned isoleucine in LA3IK and CVIL motif in RP-7 were crucial for inhibiting breast cancer cell migration. This is evident from the limited migration inhibition observed when MDA-MB-231 cells were treated with scrambled peptides LA3IK SCR and RP-7 SCR. Additionally, LA3IK and RP-7 effectively suppressed tumor growth in an orthotopic breast cancer model. Notably, mice tolerated high intraperitoneal (ip) peptide doses of 90 mg/Kg well, surpassing significantly lower doses of 5 mg/Kg intravenously (iv) and 30 mg/Kg intraperitoneally (ip) used in both in vivo pharmacokinetic studies and orthotopic mouse model assays. D-isomers of LA3IK and RP-7 showed enhanced anticancer activity compared to their L-isomers. D-LA3IK remained stable in mouse plasma for 24 h with 75% remaining, exhibiting superior pharmacokinetic properties over D/L-RP-7. In summary, our findings mark the first report of short peptides based on MIEN1 protein sequence capable of inhibiting cancer signaling pathways, effectively impeding cancer progression both in vitro and in vivo.
Collapse
Affiliation(s)
- Amit K Tripathi
- Department of Microbiology, Immunology and Genetics, University of North Texas Health Science Center, Fort Worth, Texas, USA.
| | - Priyanka P Desai
- Department of Microbiology, Immunology and Genetics, University of North Texas Health Science Center, Fort Worth, Texas, USA
| | - Antariksh Tyagi
- Yale Center for Genome Analysis (YCGA), Yale School of Medicine, New Haven, Connecticut, USA
| | - Jana B Lampe
- Department of Microbiology, Immunology and Genetics, University of North Texas Health Science Center, Fort Worth, Texas, USA
| | - Yogesh Srivastava
- Department of Genetics, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Michael Donkor
- Department of Microbiology, Immunology and Genetics, University of North Texas Health Science Center, Fort Worth, Texas, USA
| | - Harlan P Jones
- Department of Microbiology, Immunology and Genetics, University of North Texas Health Science Center, Fort Worth, Texas, USA
| | - Sergei V Dzyuba
- Department of Chemistry and Biochemistry, Texas Christian University, Fort Worth, Texas, USA
| | - Eric Crossley
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Noelle S Williams
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Jamboor K Vishwanatha
- Department of Microbiology, Immunology and Genetics, University of North Texas Health Science Center, Fort Worth, Texas, USA.
| |
Collapse
|
5
|
Misra G, Qaisar S, Singh P. CRISPR-based therapeutic targeting of signaling pathways in breast cancer. Biochim Biophys Acta Mol Basis Dis 2024; 1870:166872. [PMID: 37666438 DOI: 10.1016/j.bbadis.2023.166872] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 08/24/2023] [Accepted: 09/01/2023] [Indexed: 09/06/2023]
Abstract
Breast cancer remains a leading cause of death for women worldwide, and new treatment strategies are needed. There are innumerable anomalous genes that are responsible for the multi-factorial carcinogenesis pathway. Although several disease-causing mutations have been detected, therapy frequently focuses on attenuating the manifestation of the disease rather than harmonizing the mutation in the target area. The advent of CRISPR-Cas9 technology has revolutionized genome editing, allowing for precise and efficient manipulation of gene expression. The purpose of this review paper is to summarize recent progress in the use of CRISPR-based approaches to target key signaling pathways associated with breast cancer progression. The first section introduces basic concepts of CRISPR technology, focusing on its application in genome editing and transcriptional regulation followed by an overview of aspects involving complex signaling pathways in breast cancer such as P13K/AKT/mTOR, EPK/MAPK and Wnt/β catenin. An extensive literature search using PubMed and Google Scholar is performed for information retrieval. Further, the role of CRISPR-based interventions in regulating gene expression revealed, altered pathway activity and potential therapeutic consequences are discussed. This review will be a valuable addition to providing comprehensive knowledge of CRISPR-Cas-mediated therapeutic targeting in breast cancer.
Collapse
Affiliation(s)
- Gauri Misra
- National Institute of Biologicals, Noida 201309, UP, India.
| | - Sidra Qaisar
- National Institute of Biologicals, Noida 201309, UP, India
| | | |
Collapse
|
6
|
Wang C, Zhang Y, Zhang T, Xu J, Yan S, Liang B, Xing D. Epidermal growth factor receptor dual-target inhibitors as a novel therapy for cancer: A review. Int J Biol Macromol 2023; 253:127440. [PMID: 37839594 DOI: 10.1016/j.ijbiomac.2023.127440] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 10/10/2023] [Accepted: 10/12/2023] [Indexed: 10/17/2023]
Abstract
Overexpression of the epidermal growth factor receptor (EGFR) has been linked to several human cancers, including esophageal cancer, pancreatic cancer, anal cancer, breast cancer, and lung cancer, particularly non-small cell lung cancer (NSCLC). Therefore, EGFR has emerged as a critical target for treating solid tumors. Many 1st-, 2nd-, 3rd-, and 4th-generation EGFR single-target inhibitors with clinical efficacy have been designed and synthesized in recent years. Drug resistance caused by EGFR mutations has posed a significant challenge to the large-scale clinical application of EGFR single-target inhibitors and the discovery of novel EGFR inhibitors. Therapeutic methods for overcoming multipoint EGFR mutations are still needed in medicine. EGFR dual-target inhibitors are more promising than single-target inhibitors as they have a lower risk of drug resistance, higher efficacy, lower dosage, and fewer adverse events. EGFR dual-target inhibitors have been developed sequentially to date, providing new options for remission in patients with previously untreatable malignancies and laying the groundwork for a future generation of compounds. This paper introduces the EGFR family proteins and their synergistic effects with other anticancer targets, and provides a comprehensive review of the development of EGFR dual-target inhibitors in cancer, as well as the opportunities and challenges associated with those fields.
Collapse
Affiliation(s)
- Chao Wang
- The Affiliated Hospital of Qingdao University, Cancer Institute, Qingdao University, Qingdao 266071, Shandong, China; Qingdao Cancer Institute, Qingdao University, Qingdao 266071, Shandong, China
| | - Yujing Zhang
- The Affiliated Cardiovascular Hospital of Qingdao University, Qingdao University, Qingdao 266071, Shandong, China.
| | - Tingting Zhang
- The Affiliated Hospital of Qingdao University, Cancer Institute, Qingdao University, Qingdao 266071, Shandong, China; Qingdao Cancer Institute, Qingdao University, Qingdao 266071, Shandong, China
| | - Jiazhen Xu
- The Affiliated Hospital of Qingdao University, Cancer Institute, Qingdao University, Qingdao 266071, Shandong, China; Qingdao Cancer Institute, Qingdao University, Qingdao 266071, Shandong, China
| | - Saisai Yan
- The Affiliated Hospital of Qingdao University, Cancer Institute, Qingdao University, Qingdao 266071, Shandong, China; Qingdao Cancer Institute, Qingdao University, Qingdao 266071, Shandong, China.
| | - Bing Liang
- The Affiliated Hospital of Qingdao University, Cancer Institute, Qingdao University, Qingdao 266071, Shandong, China; Qingdao Cancer Institute, Qingdao University, Qingdao 266071, Shandong, China.
| | - Dongming Xing
- The Affiliated Hospital of Qingdao University, Cancer Institute, Qingdao University, Qingdao 266071, Shandong, China; Qingdao Cancer Institute, Qingdao University, Qingdao 266071, Shandong, China; School of Life Sciences, Tsinghua University, Beijing 100084, China
| |
Collapse
|
7
|
Buonvino S, Arciero I, Martinelli E, Seliktar D, Melino S. Modelling the disease: H 2S-sensitivity and drug-resistance of triple negative breast cancer cells can be modulated by embedding in isotropic micro-environment. Mater Today Bio 2023; 23:100862. [PMID: 38046276 PMCID: PMC10689286 DOI: 10.1016/j.mtbio.2023.100862] [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: 07/27/2023] [Revised: 09/26/2023] [Accepted: 11/09/2023] [Indexed: 12/05/2023] Open
Abstract
Three-dimensional (3D) cell culture systems provide more physiologically relevant information, representing more accurately the actual microenvironment where cells reside in tissues. However, the differences between the tissue culture plate (TCP) and 3D culture systems in terms of tumour cell growth, proliferation, migration, differentiation and response to the treatment have not been fully elucidated. Tumoroid microspheres containing the MDA-MB 231 breast cancer cell line were prepared using either tunable PEG-fibrinogen (PFs) or tunable PEG-silk fibroin (PSFs) hydrogels, respectively named MDAPFs and MDAPSFs. The cancer cells in the tumoroids showed changes both in globular morphology and at the protein expression level. A decrease of both Histone H3 acetylation and cyclin D1 expression in all 3D systems, compared to the 2D cell culture, was detected in parallel to changes of the matrix stiffness. The effects of a glutathionylated garlic extract (GSGa), a slow H2S-releasing donor, were investigated on both tumoroid systems. A pro-apoptotic effect of GSGa on tumour cell growth in 2D culture was observed as opposed to a pro-proliferative effect apparent in both MDAPFs and MDAPSFs. A dedicated ad hoc 3D cell migration chip was designed and optimized for studying tumour cell invasion in a gel-in-gel configuration. An anti-cell-invasion effect of the GSGa was observed in the 2D cell culture, whereas a pro-migratory effect in both MDAPFs and MDAPSFs was observed in the 3D cell migration chip assay. An increase of cyclin D1 expression after GSGa treatment was observed in agreement with an increase of the cell invasion index. Our results suggest that the "dimensionality" and the stiffness of the 3D cell culture milieu can change the response to both the gasotransmitter H2S and doxorubicin due to differences in both H2S diffusion and changes in protein expression. Moreover, we uncovered a direct relation between the cyclin D1 expression and the stiffness of the 3D cell culture milieu, suggesting the potential causal involvement of the cyclin D1 as a bio-marker for sensitivity of the tumour cells to their matrix stiffness. Therefore, our hydrogel-based tumoroids represent a valid tunable model for studying the physically induced transdifferentiation (PiT) of cancer cells and as a more reliable and predictive in vitro screening platform to investigate the effects of anti-tumour drugs.
Collapse
Affiliation(s)
- Silvia Buonvino
- Department of Chemical Sciences and Technologies, University of Rome “Tor Vergata”, via della Ricerca Scientifica, 00133, Rome, Italy
| | - Ilaria Arciero
- Department of Chemical Sciences and Technologies, University of Rome “Tor Vergata”, via della Ricerca Scientifica, 00133, Rome, Italy
| | - Eugenio Martinelli
- Department of Electronic Engineering, University of Rome Tor Vergata, Rome, Italy
- Interdisciplinary Center for Advanced Studies on Lab-on -Chip and Organ-on-Chip Applications, University of Rome Tor Vergata, Rome, Italy
| | - Dror Seliktar
- Department of Biomedical Engineering, Technion Israel Institute of Technology, 3200003, Haifa, Israel
| | - Sonia Melino
- Department of Chemical Sciences and Technologies, University of Rome “Tor Vergata”, via della Ricerca Scientifica, 00133, Rome, Italy
- NAST Centre, University of Rome ‘Tor Vergata’, Rome, Italy
| |
Collapse
|
8
|
Duszyc K, von Pein JB, Ramnath D, Currin-Ross D, Verma S, Lim F, Sweet MJ, Schroder K, Yap AS. Apical extrusion prevents apoptosis from activating an acute inflammatory program in epithelia. Dev Cell 2023; 58:2235-2248.e6. [PMID: 37647898 DOI: 10.1016/j.devcel.2023.08.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 06/20/2023] [Accepted: 08/07/2023] [Indexed: 09/01/2023]
Abstract
Apoptosis is traditionally considered to be an immunologically silent form of cell death. Multiple mechanisms exist to ensure that apoptosis does not stimulate the immune system to cause inflammation or autoimmunity. Against this expectation, we now report that epithelia are programmed to provoke, rather than suppress, inflammation in response to apoptosis. We found that an acute inflammatory response led by neutrophils occurs in zebrafish and cell culture when apoptotic epithelial cells cannot be expelled from the monolayer by apical extrusion. This reflects an intrinsic circuit where ATP released from apoptotic cells stimulates epithelial cells in the immediate vicinity to produce interleukin-8 (IL-8). Apical extrusion therefore prevents inappropriate epithelial inflammation by physically eliminating apoptotic cells before they can activate this pro-inflammatory circuit. This carries the implication that epithelia may be predisposed to inflammation, elicited by sporadic or induced apoptosis, if apical extrusion is compromised.
Collapse
Affiliation(s)
- Kinga Duszyc
- Division of Cell and Developmental Biology, Institute for Molecular Bioscience, The University of Queensland, Brisbane 4072, Australia.
| | - Jessica B von Pein
- Division of Cell and Developmental Biology, Institute for Molecular Bioscience, The University of Queensland, Brisbane 4072, Australia
| | - Divya Ramnath
- Division of Cell and Developmental Biology, Institute for Molecular Bioscience, The University of Queensland, Brisbane 4072, Australia
| | - Denni Currin-Ross
- Division of Cell and Developmental Biology, Institute for Molecular Bioscience, The University of Queensland, Brisbane 4072, Australia
| | - Suzie Verma
- Division of Cell and Developmental Biology, Institute for Molecular Bioscience, The University of Queensland, Brisbane 4072, Australia
| | - Fayth Lim
- Division of Cell and Developmental Biology, Institute for Molecular Bioscience, The University of Queensland, Brisbane 4072, Australia
| | - Matthew J Sweet
- Division of Cell and Developmental Biology, Institute for Molecular Bioscience, The University of Queensland, Brisbane 4072, Australia
| | - Kate Schroder
- Division of Cell and Developmental Biology, Institute for Molecular Bioscience, The University of Queensland, Brisbane 4072, Australia
| | - Alpha S Yap
- Division of Cell and Developmental Biology, Institute for Molecular Bioscience, The University of Queensland, Brisbane 4072, Australia.
| |
Collapse
|
9
|
Thakur B, Saha L, Bhatia A. Relative refractoriness of breast cancer cells to tumour necrosis factor-α induced necroptosis. Clin Exp Pharmacol Physiol 2022; 49:1294-1306. [PMID: 36054417 DOI: 10.1111/1440-1681.13711] [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: 03/22/2022] [Revised: 06/22/2022] [Accepted: 08/09/2022] [Indexed: 01/31/2023]
Abstract
Necroptosis, a recently identified programmed cell death pathway, has attracted attention as an alternative route to target apoptosis-resistant cancer cells. The status of the necroptosis pathway in different subtypes of breast cancer has not been well explored. Stimulating the cells by TNF-α can trigger cell survival or death depending on the combination of downstream players involved. In this work, we attempted to induce necroptosis in them using a combination of TNF-α and Z-VAD-FMK with and without chemotherapy. Cell viability, apoptosis, and necroptosis were assessed using MTT and Annexin-V/PI assays, respectively. Gene and protein expression was analysed by qPCR and immunophenotyping. Both the cell lines were resistant to induction of cell death by necroptosis. There was no enhancement in cell death when chemotherapeutic drugs were combined with necroptosis induction. Expression studies showed reduced translational expression of key necroptosis molecules like RIP kinases and MLKL in breast cancer cells compared to positive control cell line L929. Also, cell survival molecules were expressed more in MDA-MB-231 in contrast to death pathway molecules which were expressed more in T47D cells. In this work, the two breast cancer cell lines were observed to be resistant to TNF-α induced necroptosis with or without chemotherapy. Expression of key necroptosis players revealed relative insufficiency of the molecular machinery involved in the above pathway. In our opinion this may be the cause for resistance to necroptosis and novel strategies to upregulate these molecules need to be developed to sensitize the breast cancer cells towards cell death by necroptosis.
Collapse
Affiliation(s)
- Banita Thakur
- Department of Experimental Medicine and Biotechnology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Lekha Saha
- Department of Pharmacology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Alka Bhatia
- Department of Experimental Medicine and Biotechnology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| |
Collapse
|
10
|
Song J, Qiu H, Geng X, Gou J, Zheng Y, Nie Z, Wang J, Wang M. Immunomodulatory effects of vinegar-egg juice: Potential pharmacological effects of a traditional Chinese food remedy? J Food Biochem 2022; 46:e14418. [PMID: 36161663 DOI: 10.1111/jfbc.14418] [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: 05/24/2022] [Revised: 09/01/2022] [Accepted: 09/06/2022] [Indexed: 01/13/2023]
Abstract
Thousands of years of historical practice have proven that the ancient Chinese food, vinegar-egg juice, has immune-boosting effects with the presence of many nutritional factors. However, its mechanism of action in the body remains unclear. In this research project, vinegar-egg juice was chosen to analyze its immune-enhancing effects on mice. The immune enhancing effects of egg, vinegar and vinegar egg juice on lymphocytes of mouse spleen were compared. The effects on immune function of mice were analyzed by studying the organ index, natural killer(NK) cell activity, lymphocyte transformation function and cytokine changes in immune organs after treatment with vinegar-egg juice. The mechanism of immune enhancement was speculated by analyzing the changes of total IKKα/β/IκBα/NF-κB and its phosphorylated protein kinase by Western blot. Experiments have shown that vinegar and eggs have less immune regulation than vinegar-egg juice. Vinegar-egg juice can regulate the cellular and humoral immunity of spleen lymphocytes, increase the phosphorylated kinases of IKKα/β, reduce the total protein expression of IκBα, and activate the signaling pathway of IKK/IκB/NF-κB. In addition, compared with the control group, vinegar-egg juice reduced the Firmicutes/Bacteroidetes ratio and increased the relative abundance of beneficial bacteria. Furthermore, vinegar-egg juice can raise phosphatidylserine (PS) and serotonin (5-HT) levels in the body. The results showed that the vinegar-egg juice had obvious immunomodulatory activity. It was speculated that the intake of vinegar-egg juice can increase the activity of NK cells, T lymphocytes and B lymphocytes by increasing 5-HT levels, ultimately enhancing the body's immune function. PRACTICAL APPLICATIONS: In this work, we evaluated the immune regulation of vinegar, egg and vinegar-egg juice in mice. In addition, we investigated the effects of vinegar-egg juice on gut microbiota. And combined with the composition of the vinegar-egg juice, it was found that the intake of vinegar-egg juice could increase the activity of NK cells, T lymphocytes and B lymphocytes by increasing 5-HT levels, ultimately enhancing the body's immune function. On the basis of the results of this study, we recommend vinegar-egg juice can be a potential health food to resist the epidemic and improve autoimmunity in special times of the novel coronavirus outbreak.
Collapse
Affiliation(s)
- Jia Song
- State Key Laboratory of Food Nutrition and Safety, Tianjin Engineering Research Center of Microbial Metabolism and Fermentation Process Control, College of Biotechnology, Tianjin University of Science & Technology, Tianjin, China
| | - Huirui Qiu
- State Key Laboratory of Food Nutrition and Safety, Tianjin Engineering Research Center of Microbial Metabolism and Fermentation Process Control, College of Biotechnology, Tianjin University of Science & Technology, Tianjin, China
| | - Xiaoqi Geng
- State Key Laboratory of Food Nutrition and Safety, Tianjin Engineering Research Center of Microbial Metabolism and Fermentation Process Control, College of Biotechnology, Tianjin University of Science & Technology, Tianjin, China
| | - Jia Gou
- State Key Laboratory of Food Nutrition and Safety, Tianjin Engineering Research Center of Microbial Metabolism and Fermentation Process Control, College of Biotechnology, Tianjin University of Science & Technology, Tianjin, China
| | - Yu Zheng
- State Key Laboratory of Food Nutrition and Safety, Tianjin Engineering Research Center of Microbial Metabolism and Fermentation Process Control, College of Biotechnology, Tianjin University of Science & Technology, Tianjin, China
| | - Zhiqiang Nie
- Key Laboratory of Chemical Biology and Molecular Engineering, Ministry of Education, Institute of Biotechnology, Shanxi University, Taiyuan, China
| | - Jianxin Wang
- Wisconsin Center for NanoBioSystems, School of Pharmacy, University of Wisconsin-Madison, Madison, Wisconsin, USA.,Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Min Wang
- State Key Laboratory of Food Nutrition and Safety, Tianjin Engineering Research Center of Microbial Metabolism and Fermentation Process Control, College of Biotechnology, Tianjin University of Science & Technology, Tianjin, China
| |
Collapse
|
11
|
Engineered nanoparticles as emerging gene/drug delivery systems targeting the nuclear factor-κB protein and related signaling pathways in cancer. Biomed Pharmacother 2022; 156:113932. [DOI: 10.1016/j.biopha.2022.113932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Revised: 10/25/2022] [Accepted: 10/26/2022] [Indexed: 11/06/2022] Open
|
12
|
Yi YW, You KS, Han S, Ha IJ, Park JS, Lee SG, Seong YS. Inhibition of IκB Kinase Is a Potential Therapeutic Strategy to Circumvent Resistance to Epidermal Growth Factor Receptor Inhibition in Triple-Negative Breast Cancer Cells. Cancers (Basel) 2022; 14:5215. [PMID: 36358633 PMCID: PMC9654813 DOI: 10.3390/cancers14215215] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 10/21/2022] [Accepted: 10/21/2022] [Indexed: 03/21/2024] Open
Abstract
Triple-negative breast cancer (TNBC) remains as an intractable malignancy with limited therapeutic targets. High expression of epidermal growth factor receptor (EGFR) has been associated with a poor prognosis of TNBC; however, EGFR targeting has failed with unfavorable clinical outcomes. Here, we performed a combinatorial screening of fifty-five protein kinase inhibitors with the EGFR inhibitor gefitinib in the TNBC cell line MDA-MB-231 and identified the IκB kinase (IKK) inhibitor IKK16 as a sensitizer of gefitinib. Cell viability and clonogenic survival assays were performed to evaluate the antiproliferative effects of the gefitinib and IKK16 (Gefitinib + IKK16) combination in TNBC cell lines. Western blot analyses were also performed to reveal the potential mode of action of this combination. In addition, next-generation sequencing (NGS) analysis was performed in Gefitinib+IKK16-treated cells. The Gefitinib+IKK16 treatment synergistically reduced cell viability and colony formation of TNBC cell lines such as HS578T, MDA-MB-231, and MDA-MB-468. This combination downregulated p-STAT3, p-AKT, p-mTOR, p-GSK3β, and p-RPS6. In addition, p-NF-κB and the total NF-κB were also regulated by this combination. Furthermore, NGS analysis revealed that NF-κB/RELA targets including CCL2, CXCL8, EDN1, IL-1β, IL-6, and SERPINE1 were further reduced and several potential tumor suppressors, such as FABP3, FADS2, FDFT1, SEMA6A, and PCK2, were synergistically induced by the Gefitinib-+IKK16 treatment. Taken together, we identified the IKK/NF-κB pathway as a potential target in combination of EGFR inhibition for treating TNBC.
Collapse
Affiliation(s)
- Yong Weon Yi
- Department of Biochemistry, College of Medicine, Dankook University, Cheonan 31116, Chungcheongnam-do, Korea
| | - Kyu Sic You
- Department of Biochemistry, College of Medicine, Dankook University, Cheonan 31116, Chungcheongnam-do, Korea
- Graduate School of Convergence Medical Science, Dankook University, Cheonan 31116, Chungcheongnam-do, Korea
| | - Sanghee Han
- Graduate School, Kyung Hee University, Seoul 02447, Korea
| | - In Jin Ha
- Graduate School, Kyung Hee University, Seoul 02447, Korea
| | - Jeong-Soo Park
- Department of Biochemistry, College of Medicine, Dankook University, Cheonan 31116, Chungcheongnam-do, Korea
| | - Seok-Geun Lee
- Graduate School, Kyung Hee University, Seoul 02447, Korea
| | - Yeon-Sun Seong
- Department of Biochemistry, College of Medicine, Dankook University, Cheonan 31116, Chungcheongnam-do, Korea
- Graduate School of Convergence Medical Science, Dankook University, Cheonan 31116, Chungcheongnam-do, Korea
| |
Collapse
|
13
|
Xing J, Chen W, Chen K, Zhu S, Lin F, Qi Y, Zhang Y, Han S, Rao T, Ruan Y, Zhao S, Yu W, Cheng F. TFAP2C Knockdown Sensitizes Bladder Cancer Cells to Cisplatin Treatment via Regulation of EGFR and NF-κB. Cancers (Basel) 2022; 14:cancers14194809. [PMID: 36230734 PMCID: PMC9562889 DOI: 10.3390/cancers14194809] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 09/27/2022] [Accepted: 09/28/2022] [Indexed: 11/22/2022] Open
Abstract
Simple Summary Bladder cancer (BCa) is considered one of the most common neoplasms of the urology system. Cisplatin-based chemotherapy has been the primary treatment for patients with advanced or metastatic BCa. Nevertheless, cisplatin resistance often limits the treatment of bladder cancer. We expect to find approaches to improve the therapeutic efficacy of cisplatin in bladder cancer. In recent years, many studies have shown that transcription factor AP-2 gamma (TFAP2C) acts as a key player in cancer development and and its expression level is closely related to the sensitivity of tumors to cisplatin. Our study investigated whether TFAP2C affects the sensitivity of BCa cells to cisplatin and the possible mechanisms. We found that TFAP2C expression was significantly upregulated in most BCa tissues compared to adjacent normal tissues. The present study confirmed that TFAP2C knockdown enhanced the anti-tumor effects of cisplatin by decreasing cisplatin-induced activation levels of epidermal growth factor receptor (EGFR) and nuclear factor kappaB (NF-κB). Specifically, this study provides a novel approach to improve the efficacy of cisplatin. Abstract Cisplatin is the first-line chemotherapy for advanced or metastatic bladder cancer. Nevertheless, approximately half of patients with BCa are insensitive to cisplatin therapy or develop cisplatin resistance during the treatment process. Therefore, it is especially crucial to investigate ways to enhance the sensitivity of tumor cells to cisplatin. Transcription factor AP-2 gamma (TFAP2C) is involved in cancer development and chemotherapy sensitivity. However, its relationship with chemotherapy has not been studied in BCa. In this study, we aimed to investigate the therapeutic potential of TFAP2C in human BCa. Results based on TCGA (The Cancer Genome Atlas), GTEx (The Genotype-Tissue Expression) and GEO (Gene Expression Omnibus) data showed that TFAP2C expression was upregulated in BCa tissues and that its high expression was associated with poor prognosis. Meanwhile, we demonstrated the overexpression of TFAP2C in BCa clinical specimens. Subsequently, in vitro, we knocked down TFAP2C in BCa cells and found that TFAP2C knockdown further increased cell cycle arrest and apoptosis caused by cisplatin. In addition, the inhibitory effect of cisplatin on BCa cell migration and invasion was enhanced by TFAP2C knockdown. Our data indicated that cisplatin increased epidermal growth factor receptor (EGFR) and nuclear factor-kappaB (NF-κB) activation levels, but TFAP2C knockdown suppressed this effect. Finally, in vivo data further validated these findings. Our study showed that TFAP2C knockdown affected the activation levels of EGFR and NF-κB and enhanced the anti-tumor effects of cisplatin in vivo and in vitro. This provides a new direction to improve the efficacy of traditional cisplatin chemotherapy.
Collapse
Affiliation(s)
- Ji Xing
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Wu Chen
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Kang Chen
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Shaoming Zhu
- Department of Urology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| | - Fangyou Lin
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Yucheng Qi
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Yunlong Zhang
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Shangting Han
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Ting Rao
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Yuan Ruan
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Sheng Zhao
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Weimin Yu
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Correspondence: (W.Y.); (F.C.)
| | - Fan Cheng
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Correspondence: (W.Y.); (F.C.)
| |
Collapse
|
14
|
Wang R, Wen ZY, Liu FH, Wei YF, Xu HL, Sun ML, Zhao YH, Gong TT, Wang HH, Wu QJ. Association between dietary acid load and cancer risk and prognosis: An updated systematic review and meta-analysis of observational studies. Front Nutr 2022; 9:891936. [PMID: 35967803 PMCID: PMC9365077 DOI: 10.3389/fnut.2022.891936] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 07/04/2022] [Indexed: 11/13/2022] Open
Abstract
Epidemiological studies have suggested that dietary acid load (DAL) might be related to the risk and prognosis of cancer, whereas the evidence is contentious. Several high-quality observational studies have been published following a prior systematic review with only one study included. Consequently, we conducted an updated systematic review and meta-analysis to comprehensively investigate the relationship between DAL and cancer risk and prognosis. A systematic literature search was conducted in the PubMed, Embase, and Web of Science databases from inception to 26 October 2021. Summary relative risks (RRs) with 95% CIs were calculated using a random-effects model. Publication bias, subgroup, meta-regression, and sensitivity analyses were also conducted. Ten observational studies (six cohorts and four case–control studies) with 227,253 participants were included in this systematic review and meta-analysis. The summary RRs revealed a statistically significant associations between DAL and cancer risk (RR = 1.58, 95% CI = 1.23–2.05, I2 = 71.9%, n = 7) and prognosis (RR = 1.53, 95% CI = 1.10–2.13, I2 = 77.1%, n = 3). No evidence of publication bias was observed in the current analysis. Positive associations were observed in most subgroup analyses stratified by predefined factors, including region, study design, study quality, study population, participants’ gender, age of participants, cancer type, DAL assessment indicator, and adjustment of potential confounding parameters. No evidence of heterogeneity between subgroups was indicated by meta-regression analyses. The high DAL might be associated with an increased risk of cancer, as well as a poor prognosis of cancer. More high-quality prospective studies are warranted to further determine the associations between DAL and risk and prognosis for specific cancers.
Collapse
Affiliation(s)
- Ran Wang
- Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, Shenyang, China.,Clinical Research Center, Shengjing Hospital of China Medical University, Shenyang, China
| | - Zhao-Yan Wen
- Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, Shenyang, China.,Clinical Research Center, Shengjing Hospital of China Medical University, Shenyang, China
| | - Fang-Hua Liu
- Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, Shenyang, China.,Clinical Research Center, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yi-Fan Wei
- Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, Shenyang, China.,Clinical Research Center, Shengjing Hospital of China Medical University, Shenyang, China
| | - He-Li Xu
- Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, Shenyang, China.,Clinical Research Center, Shengjing Hospital of China Medical University, Shenyang, China
| | - Ming-Li Sun
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yu-Hong Zhao
- Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, Shenyang, China.,Clinical Research Center, Shengjing Hospital of China Medical University, Shenyang, China
| | - Ting-Ting Gong
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Hui-Han Wang
- Department of Hematology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Qi-Jun Wu
- Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, Shenyang, China.,Clinical Research Center, Shengjing Hospital of China Medical University, Shenyang, China.,Department of Hematology, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
15
|
Revisiting Epithelial Carcinogenesis. Int J Mol Sci 2022; 23:ijms23137437. [PMID: 35806442 PMCID: PMC9267463 DOI: 10.3390/ijms23137437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 06/22/2022] [Accepted: 06/22/2022] [Indexed: 12/04/2022] Open
Abstract
The origin of cancer remains one of the most important enigmas in modern biology. This paper presents a hypothesis for the origin of carcinomas in which cellular aging and inflammation enable the recovery of cellular plasticity, which may ultimately result in cancer. The hypothesis describes carcinogenesis as the result of the dedifferentiation undergone by epithelial cells in hyperplasia due to replicative senescence towards a mesenchymal cell state with potentially cancerous behavior. In support of this hypothesis, the molecular, cellular, and histopathological evidence was critically reviewed and reinterpreted when necessary to postulate a plausible generic series of mechanisms for the origin and progression of carcinomas. In addition, the implications of this theoretical framework for the current strategies of cancer treatment are discussed considering recent evidence of the molecular events underlying the epigenetic switches involved in the resistance of breast carcinomas. The hypothesis also proposes an epigenetic landscape for their progression and a potential mechanism for restraining the degree of dedifferentiation and malignant behavior. In addition, the manuscript revisits the gradual degeneration of the nonalcoholic fatty liver disease to propose an integrative generalized mechanistic explanation for the involution and carcinogenesis of tissues associated with aging. The presented hypothesis might serve to understand and structure new findings into a more encompassing view of the genesis of degenerative diseases and may inspire novel approaches for their study and therapy.
Collapse
|
16
|
Tan L, Zhang J, Wang Y, Wang X, Wang Y, Zhang Z, Shuai W, Wang G, Chen J, Wang C, Ouyang L, Li W. Development of Dual Inhibitors Targeting Epidermal Growth Factor Receptor in Cancer Therapy. J Med Chem 2022; 65:5149-5183. [PMID: 35311289 DOI: 10.1021/acs.jmedchem.1c01714] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Epidermal growth factor receptor (EGFR) is of great significance in mediating cell signaling transduction and tumor behaviors. Currently, third-generation inhibitors of EGFR, especially osimertinib, are at the clinical frontier for the treatment of EGFR-mutant non-small-cell lung cancer (NSCLC). Regrettably, the rapidly developing drug resistance caused by EGFR mutations and the compensatory mechanism have largely limited their clinical efficacy. Given the synergistic effect between EGFR and other compensatory targets during tumorigenesis and tumor development, EGFR dual-target inhibitors are promising for their reduced risk of drug resistance, higher efficacy, lower dosage, and fewer adverse events than those of single-target inhibitors. Hence, we present the synergistic mechanism underlying the role of EGFR dual-target inhibitors against drug resistance, their structure-activity relationships, and their therapeutic potential. Most importantly, we emphasize the optimal target combinations and design strategies for EGFR dual-target inhibitors and provide some perspectives on new challenges and future directions in this field.
Collapse
Affiliation(s)
- Lun Tan
- Department of Respiratory and Critical Care Medicine, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041 Sichuan, China.,Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, 610041 Sichuan, China.,Precision Medicine Key Laboratory of Sichuan Province and Precision Medicine Research Center, West China Hospital, Sichuan University, Chengdu, 610041 Sichuan, China
| | - Jifa Zhang
- Department of Respiratory and Critical Care Medicine, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041 Sichuan, China.,Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, 610041 Sichuan, China.,Precision Medicine Key Laboratory of Sichuan Province and Precision Medicine Research Center, West China Hospital, Sichuan University, Chengdu, 610041 Sichuan, China
| | - Yuxi Wang
- Department of Respiratory and Critical Care Medicine, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041 Sichuan, China.,Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, 610041 Sichuan, China.,Precision Medicine Key Laboratory of Sichuan Province and Precision Medicine Research Center, West China Hospital, Sichuan University, Chengdu, 610041 Sichuan, China
| | - Xiye Wang
- Department of Respiratory and Critical Care Medicine, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041 Sichuan, China
| | - Yanyan Wang
- Department of Respiratory and Critical Care Medicine, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041 Sichuan, China
| | - Zhixiong Zhang
- Department of Respiratory and Critical Care Medicine, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041 Sichuan, China
| | - Wen Shuai
- Department of Respiratory and Critical Care Medicine, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041 Sichuan, China.,Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, 610041 Sichuan, China.,Precision Medicine Key Laboratory of Sichuan Province and Precision Medicine Research Center, West China Hospital, Sichuan University, Chengdu, 610041 Sichuan, China
| | - Guan Wang
- Department of Respiratory and Critical Care Medicine, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041 Sichuan, China
| | - Juncheng Chen
- Department of Respiratory and Critical Care Medicine, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041 Sichuan, China.,Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, 610041 Sichuan, China.,Precision Medicine Key Laboratory of Sichuan Province and Precision Medicine Research Center, West China Hospital, Sichuan University, Chengdu, 610041 Sichuan, China
| | - Chengdi Wang
- Department of Respiratory and Critical Care Medicine, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041 Sichuan, China.,Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, 610041 Sichuan, China
| | - Liang Ouyang
- Department of Respiratory and Critical Care Medicine, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041 Sichuan, China.,Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, 610041 Sichuan, China.,Precision Medicine Key Laboratory of Sichuan Province and Precision Medicine Research Center, West China Hospital, Sichuan University, Chengdu, 610041 Sichuan, China
| | - Weimin Li
- Department of Respiratory and Critical Care Medicine, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041 Sichuan, China.,Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, 610041 Sichuan, China.,Precision Medicine Key Laboratory of Sichuan Province and Precision Medicine Research Center, West China Hospital, Sichuan University, Chengdu, 610041 Sichuan, China
| |
Collapse
|
17
|
Mazurek M, Szczepanek D, Orzyłowska A, Rola R. Analysis of Factors Affecting 5-ALA Fluorescence Intensity in Visualizing Glial Tumor Cells-Literature Review. Int J Mol Sci 2022; 23:ijms23020926. [PMID: 35055109 PMCID: PMC8779265 DOI: 10.3390/ijms23020926] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 01/11/2022] [Accepted: 01/13/2022] [Indexed: 01/27/2023] Open
Abstract
Glial tumors are one of the most common lesions of the central nervous system. Despite the implementation of appropriate treatment, the prognosis is not successful. As shown in the literature, maximal tumor resection is a key element in improving therapeutic outcome. One of the methods to achieve it is the use of fluorescent intraoperative navigation with 5-aminolevulinic acid. Unfortunately, often the level of fluorescence emitted is not satisfactory, resulting in difficulties in the course of surgery. This article summarizes currently available knowledge regarding differences in the level of emitted fluorescence. It may depend on both the histological type and the genetic profile of the tumor, which is reflected in the activity and expression of enzymes involved in the intracellular metabolism of fluorescent dyes, such as PBGD, FECH, UROS, and ALAS. The transport of 5-aminolevulinic acid and its metabolites across the blood–brain barrier and cell membranes mediated by transporters, such as ABCB6 and ABCG2, is also important. Accompanying therapies, such as antiepileptic drugs or steroids, also have an impact on light emission by tumor cells. Accurate determination of the factors influencing the fluorescence of 5-aminolevulinic acid-treated cells may contribute to the improvement of fluorescence navigation in patients with highly malignant gliomas.
Collapse
|
18
|
Hu L, Fan M, Shi S, Song X, Wang F, He H, Qi B. Dual target inhibitors based on EGFR: Promising anticancer agents for the treatment of cancers (2017-). Eur J Med Chem 2022; 227:113963. [PMID: 34749202 DOI: 10.1016/j.ejmech.2021.113963] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 10/23/2021] [Accepted: 10/28/2021] [Indexed: 02/06/2023]
Abstract
The EGFR family play a significant role in cell signal transduction and their overexpression is implicated in the pathogenesis of numerous human solid cancers. Inhibition of the EGFR-mediated signaling pathways by EGFR inhibitors is a widely used strategy for the treatment of cancers. In most cases, the EGFR inhibitors used in clinic were only effective when the cancer cells harbored specific activating EGFR mutations which appeared to preserve the ligand-dependency of receptor activation but altered the pattern of downstream signaling pathways. Moreover, cancer is a kind of multifactorial disease, and therefore manipulating a single target may result in treatment failure. Although drug combinations for the treatment of cancers proved to be successful, the use of two or more drugs concurrently still was a challenge in clinical therapy owing to various dose-limiting toxicities and drug-drug interactions caused by pharmacokinetic profiles changed. Therefore, a single drug targeting two or multiple targets could serve as an effective strategy for the treatment of cancers. In recent, drugs with diverse pharmacological effects have been shown to be more advantageous than combination therapies due to their lower incidences of side effects and more resilient therapies. Accordingly, dual target-single-agent strategy has become a popular field for cancer treatment, and researchers became more and more interest in the development of novel dual-target drugs in recent years. In this review, we briefly introduce the EGFR family proteins and synergisms between EGFR and other anticancer targets, and summarizes the development of potential dual target inhibitors based on wild-type and/or mutant EGFR for the treatment of solid cancers in the past five years. Additionally, the rational design and SARs of these dual target agents are also presented in detailed, which will lay a significant foundation for the further development of novel EGFR-based dual inhibitors with excellent druggability.
Collapse
Affiliation(s)
- Liping Hu
- Department of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai, 519041, Guangdong Province, China
| | - Mengmeng Fan
- Department of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai, 519041, Guangdong Province, China
| | - Shengmin Shi
- Department of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai, 519041, Guangdong Province, China
| | - Xiaomeng Song
- Department of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai, 519041, Guangdong Province, China
| | - Fei Wang
- Department of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai, 519041, Guangdong Province, China
| | - Huan He
- Department of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai, 519041, Guangdong Province, China.
| | - Baohui Qi
- Department of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai, 519041, Guangdong Province, China.
| |
Collapse
|
19
|
Abstract
Tomentosin is a natural compound known for its presence in some medicinal plants of the Asteraceae family such as Inula viscosa. Recent studies have highlighted its anticancer and anti-inflammatory properties. Its anticancer mechanisms are unique and act at different levels ranging from cellular organization to molecular transcriptional factors and epigenetic modifications. Tomentosin’s possession of the modulatory effect on telomerase expression on tumor cell lines has captured the interest of researchers and spurred a more robust study on its anticancer effect. Since inflammation has a close link with cancer disease, this natural compound appears to be a potential cancer-fighting drug. Indeed, its recently demonstrated anti-inflammatory action can be considered as a starting point for its evaluation as an anticancer chemo-preventive agent
Collapse
|
20
|
Park YMM, Shivappa N, Petimar J, Hodgson ME, Nichols HB, Steck SE, Hébert JR, Sandler DP. Dietary inflammatory potential, oxidative balance score, and risk of breast cancer: Findings from the Sister Study. Int J Cancer 2021; 149:615-626. [PMID: 33783833 PMCID: PMC8256885 DOI: 10.1002/ijc.33581] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 02/09/2021] [Accepted: 03/04/2021] [Indexed: 12/16/2022]
Abstract
Diet, inflammation, and oxidative stress may be important in breast carcinogenesis, but evidence on the role of the inflammatory and prooxidative potential of dietary patterns is limited. Energy adjusted-Dietary Inflammatory Index (E-DII™) and dietary oxidative balance score (D-OBS) were calculated for 43 563 Sister Study cohort participants who completed a Block 1998 food frequency questionnaire at enrollment in 2003-2009 and satisfied eligibility criteria. D-OBS was validated using measured F2 -isoprostanes and metabolites. High E-DII score and low D-OBS represent a more proinflammatory and prooxidant diet, respectively, and associations of quartiles of each index with breast cancer (BC) risk were estimated using multivariable Cox proportional hazards regression. There were 2619 BCs diagnosed at least 1 year after enrollment (mean follow-up 8.4 years). There was no overall association between E-DII and BC risk, whereas there was a suggestive inverse association for the highest vs lowest quartile of D-OBS (HR 0.92 [95% CI, 0.81-1.03]). The highest quartile of E-DII was associated with risk of triple-negative BC (HR 1.53 [95% CI, 0.99-2.35]). When the two indices were combined, a proinflammatory/prooxidant diet (highest tertile of E-DII and lowest tertile of D-OBS) was associated with increased risk for all BC (HR 1.13 [95% CI, 1.00-1.27]) and for triple-negative BC (1.72 [95% CI, 1.10-2.70]), compared to an antiinflammatory/antioxidant diet (lowest tertile of E-DII and highest tertile of D-OBS). Diets with increased inflammatory potential and reduced oxidative balance were positively associated with overall and triple-negative BC.
Collapse
Affiliation(s)
- Yong-Moon Mark Park
- Epidemiology Branch, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, USA
- Department of Epidemiology, Fay W. Boozman College of Public Health, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Nitin Shivappa
- Cancer Prevention and Control Program, University of South Carolina, Columbia, South Carolina, USA
- Department of Epidemiology and Biostatistics, Arnold School of Public Health, University of South Carolina, Columbia, South Carolina, USA
- Connecting Health Innovations, LLC, Columbia, South Carolina, USA
| | - Joshua Petimar
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
- Department of Population Medicine, Harvard Pilgrim Health Care Institute and Harvard Medical School, Boston, Massachusetts, USA
| | | | - Hazel B Nichols
- Department of Epidemiology, University of North Carolina Gillings School of Global Public Health, Chapel Hill, North Carolina, USA
| | - Susan E Steck
- Cancer Prevention and Control Program, University of South Carolina, Columbia, South Carolina, USA
- Department of Epidemiology and Biostatistics, Arnold School of Public Health, University of South Carolina, Columbia, South Carolina, USA
| | - James R Hébert
- Cancer Prevention and Control Program, University of South Carolina, Columbia, South Carolina, USA
- Department of Epidemiology and Biostatistics, Arnold School of Public Health, University of South Carolina, Columbia, South Carolina, USA
- Connecting Health Innovations, LLC, Columbia, South Carolina, USA
| | - Dale P Sandler
- Epidemiology Branch, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, USA
| |
Collapse
|
21
|
Fruit and vegetable consumption and incident breast cancer: a systematic review and meta-analysis of prospective studies. Br J Cancer 2021; 125:284-298. [PMID: 34006925 PMCID: PMC8292326 DOI: 10.1038/s41416-021-01373-2] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 02/22/2021] [Accepted: 03/16/2021] [Indexed: 02/01/2023] Open
Abstract
BACKGROUND We conducted a systematic review and meta-analysis of prospective studies to clarify the relation of fruit and vegetable consumption with incident breast cancer. METHODS We searched systematically PubMed and EMBASE databases up to November 2020 to include prospective studies that reported the association of fruit and vegetable consumption with incident breast cancer. The pooled relative risks (RRs) and 95% confidence intervals (CIs) were calculated for the highest versus the lowest category of total fruit and vegetable, total fruit and total vegetable consumption, as well as fruit juice and subgroups of vegetables in relation to breast cancer incidence, using a random-effect model. RESULTS Total fruit and vegetable consumption was associated with lower overall (RR = 0.91, 95% CI = 0.87-0.95) and postmenopausal breast cancer risk (RR = 0.88, 95% CI = 0.79-0.99). Total fruit consumption was associated with lower overall (RR = 0.93, 95% CI = 0.88-0.99) and postmenopausal breast cancer risk (RR = 0.93, 95% CI = 0.87-0.99). Total fruit and vegetable intake were associated with 11% and 26% lower risk of oestrogen- and progesterone-receptor-positive (ER+/PR+) and -negative (ER-/PR-) breast cancer, respectively. Total vegetable consumption was associated with 27% lower risk of ER-/PR- breast cancer. Fruit juice consumption was associated with increased overall breast cancer risk (RR = 1.04, 95% CI = 1.01-1.07). We did not find significant associations for subgroups of vegetable intake and breast cancer risk. CONCLUSIONS These findings suggest that high total fruit and vegetable consumption are associated with reduced risk of overall, postmenopausal, ER+/PR+ and ER-/PR- breast cancer.
Collapse
|
22
|
Leesakul N, Kullawanichaiyanan K, Mutić S, Guzsvány V, Nhukeaw T, Ratanaphan A, Saithong S, Konno T, Sirimahachai U, Promarak V. A photoactive iridium(III) complex with 3-methyl-2-phenyl pyridine and 1,1-bis(diphenylphosphino)methane: Synthesis, structural characterization and cytotoxicity in breast cancer cells. J COORD CHEM 2021. [DOI: 10.1080/00958972.2021.1949585] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Affiliation(s)
- Nararak Leesakul
- Division of Physical Science and Center of Excellence for Innovation in Chemistry, Faculty of Science, Prince of Songkla University, Hat-Yai, Thailand
| | - Keerati Kullawanichaiyanan
- Division of Physical Science and Center of Excellence for Innovation in Chemistry, Faculty of Science, Prince of Songkla University, Hat-Yai, Thailand
| | - Sanja Mutić
- Department of Chemistry, Biochemistry and Environmental Protection, Faculty of Sciences, University of Novi Sad, Novi Sad, Serbia
| | - Valéria Guzsvány
- Department of Chemistry, Biochemistry and Environmental Protection, Faculty of Sciences, University of Novi Sad, Novi Sad, Serbia
| | - Tidarat Nhukeaw
- Department of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences, Prince of Songkla University, Hat Yai, Thailand
| | - Adisorn Ratanaphan
- Department of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences, Prince of Songkla University, Hat Yai, Thailand
| | - Saowanit Saithong
- Division of Physical Science and Center of Excellence for Innovation in Chemistry, Faculty of Science, Prince of Songkla University, Hat-Yai, Thailand
| | - Takumi Konno
- Department of Chemistry, Graduate School of Science, Osaka University, Suita, Japan
| | - Uraiwan Sirimahachai
- Division of Physical Science and Center of Excellence for Innovation in Chemistry, Faculty of Science, Prince of Songkla University, Hat-Yai, Thailand
| | - Vinich Promarak
- Department Materials Science and Engineering, School of Molecular Science and Engineering, Vidyasirimedhi Institute of Science and Technology, Wangchan, Thailand
| |
Collapse
|
23
|
You KS, Yi YW, Cho J, Park JS, Seong YS. Potentiating Therapeutic Effects of Epidermal Growth Factor Receptor Inhibition in Triple-Negative Breast Cancer. Pharmaceuticals (Basel) 2021; 14:589. [PMID: 34207383 PMCID: PMC8233743 DOI: 10.3390/ph14060589] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 06/07/2021] [Accepted: 06/14/2021] [Indexed: 12/13/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is a subset of breast cancer with aggressive characteristics and few therapeutic options. The lack of an appropriate therapeutic target is a challenging issue in treating TNBC. Although a high level expression of epidermal growth factor receptor (EGFR) has been associated with a poor prognosis among patients with TNBC, targeted anti-EGFR therapies have demonstrated limited efficacy for TNBC treatment in both clinical and preclinical settings. However, with the advantage of a number of clinically approved EGFR inhibitors (EGFRis), combination strategies have been explored as a promising approach to overcome the intrinsic resistance of TNBC to EGFRis. In this review, we analyzed the literature on the combination of EGFRis with other molecularly targeted therapeutics or conventional chemotherapeutics to understand the current knowledge and to provide potential therapeutic options for TNBC treatment.
Collapse
Affiliation(s)
- Kyu Sic You
- Department of Biochemistry, College of Medicine, Dankook University, Cheonan 31116, Chungcheongnam-do, Korea;
- Graduate School of Convergence Medical Science, Dankook University, Cheonan 3116, Chungcheongnam-do, Korea
| | - Yong Weon Yi
- Department of Nanobiomedical Science, Dankook University, Cheonan 31116, Chungcheongnam-do, Korea; (Y.W.Y.); (J.C.)
| | - Jeonghee Cho
- Department of Nanobiomedical Science, Dankook University, Cheonan 31116, Chungcheongnam-do, Korea; (Y.W.Y.); (J.C.)
| | - Jeong-Soo Park
- Department of Biochemistry, College of Medicine, Dankook University, Cheonan 31116, Chungcheongnam-do, Korea;
| | - Yeon-Sun Seong
- Department of Biochemistry, College of Medicine, Dankook University, Cheonan 31116, Chungcheongnam-do, Korea;
- Graduate School of Convergence Medical Science, Dankook University, Cheonan 3116, Chungcheongnam-do, Korea
- Department of Nanobiomedical Science, Dankook University, Cheonan 31116, Chungcheongnam-do, Korea; (Y.W.Y.); (J.C.)
| |
Collapse
|
24
|
Fan Q, An Z, Wong RA, Luo X, Lu ED, Baldwin A, Mayekar MK, Haderk F, Shokat KM, Bivona TG, Weiss WA. Betacellulin drives therapy resistance in glioblastoma. Neuro Oncol 2021; 22:457-469. [PMID: 31678994 DOI: 10.1093/neuonc/noz206] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND The transcription factor signal transducer and activator of transcription 3 (STAT3) drives progression in glioblastoma (GBM), suggesting STAT3 as a therapeutic target. Surprisingly however, GBM cells generally show primary resistance to STAT3 blockade. METHODS Human glioblastoma cell lines LN229, U87, SF767, and U373, and patient-derived xenografts (PDXs) GBM8 and GBM43 were used to evaluate epidermal growth factor receptor (EGFR) activation during STAT3 inhibition. Protein and gene expression experiments, protein stability assays, cytokine arrays, phospho-tyrosine arrays and EGFR-ligand protein arrays were performed on STAT3 inhibitor-treated cells. To evaluate antitumor activity, we administered a betacellulin (BTC)-neutralizing antibody alone and in combination with STAT3 inhibition. BTC is an EGFR ligand. We therefore treated mice with orthotopic xenografts using the third-generation EGFR inhibitor osimertinib, with or without STAT3 knockdown. RESULTS We demonstrate that both small-molecule inhibitors and knockdown of STAT3 led to expression and secretion of the EGFR ligand BTC, resulting in activation of EGFR and subsequent downstream phosphorylation of nuclear factor-kappaB (NF-κB). Neutralizing antibody against BTC abrogated activation of both EGFR and NF-κB in response to inhibition of STAT3; with combinatorial blockade of STAT3 and BTC inducing apoptosis in GBM cells. Blocking EGFR and STAT3 together inhibited tumor growth, improving survival in mice bearing orthotopic GBM PDXs in vivo. CONCLUSION These data reveal a feedback loop among STAT3, EGFR, and NF-κB that mediates primary resistance to STAT3 blockade and suggest strategies for therapeutic intervention.
Collapse
Affiliation(s)
- Qiwen Fan
- Department of Neurology, University of California San Francisco (UCSF), San Francisco, California.,Helen Diller Family Comprehensive Cancer Center, San Francisco, California
| | - Zhenyi An
- Department of Neurology, University of California San Francisco (UCSF), San Francisco, California.,Helen Diller Family Comprehensive Cancer Center, San Francisco, California
| | - Robyn A Wong
- Department of Neurology, University of California San Francisco (UCSF), San Francisco, California.,Helen Diller Family Comprehensive Cancer Center, San Francisco, California
| | - Xujun Luo
- Department of Neurology, University of California San Francisco (UCSF), San Francisco, California.,Helen Diller Family Comprehensive Cancer Center, San Francisco, California
| | - Edbert D Lu
- Department of Neurology, University of California San Francisco (UCSF), San Francisco, California.,Helen Diller Family Comprehensive Cancer Center, San Francisco, California
| | - Albert Baldwin
- Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Manasi K Mayekar
- Helen Diller Family Comprehensive Cancer Center, San Francisco, California
| | - Franziska Haderk
- Helen Diller Family Comprehensive Cancer Center, San Francisco, California
| | - Kevan M Shokat
- Howard Hughes Medical Institute and Department of Cellular and Molecular Pharmacology, University of California San Francisco, San Francisco, California
| | - Trever G Bivona
- Helen Diller Family Comprehensive Cancer Center, San Francisco, California
| | - William A Weiss
- Department of Neurology, University of California San Francisco (UCSF), San Francisco, California.,Helen Diller Family Comprehensive Cancer Center, San Francisco, California.,Department of Pediatrics, University of California San Francisco, San Francisco, California.,Department of Neurological Surgery, University of California San Francisco, San Francisco, California
| |
Collapse
|
25
|
Zhang S, Luo T, Wang J. Stable Cells with NF-κB-ZsGreen Fused Genes Created by TALEN Editing and Homology Directed Repair for Screening Anti-inflammation Drugs. J Inflamm Res 2021; 14:917-928. [PMID: 33762839 PMCID: PMC7982563 DOI: 10.2147/jir.s298938] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Accepted: 02/22/2021] [Indexed: 12/15/2022] Open
Abstract
Background NF-κB is a sequence-specific DNA-binding transcription factor that plays key roles in inflammation and cancer. It is well known that NF-κB is over-activated in these diseases. NF-κB inhibitors are therefore developed as promising drugs for these diseases. However, finding NF-κB inhibitors is dependent on effective screening platforms. Methods For providing an easy and visualizable tool for screening NF-κB inhibitors, and other NF-κB-related studies, this study edited all five genes of NF-κB family (RELA, RELB, CREL, NF-κB1, NF-κB2) in three different cell lines (293T, HepG2, and PANC1) with both TALEN and CRISPR. The edited NF-κB genes were repaired by homology-dependent repair using a linear homologous donor containing ZsGreen coding sequence. The edit efficiency was thus directly evaluated by detecting cellular fluorescence. The editing efficiency was also confirmed by PCR detection of NF-κB-ZsGreen fused genes. Results It was found that all genes were more efficiently edited by TALEN in all cells than CRISPR. The positive cells were then isolated from the TALEN-edited cell pool by flow cytometry. The purified positive cells were finally evaluated by regulating NF-κB activity with a known NF-κB inhibitor, BAY 11-7082, and an NF-κB-targeting artificial microRNA, miR533. The results revealed that all the labeled NF-κB genes responded well to the two kinds of NF-κB activity regulators in all cell lines. Conclusion This study thus obtained 15 cell lines with NF-κB-ZsGreen fused genes, which provide an easy and visualizable tool for screening NF-κB inhibitors and other NF-κB-related studies.
Collapse
Affiliation(s)
- Shuyan Zhang
- State Key Laboratory of Bioelectronics, Southeast University, Nanjing, 210096, People's Republic of China
| | - Tao Luo
- State Key Laboratory of Bioelectronics, Southeast University, Nanjing, 210096, People's Republic of China
| | - Jinke Wang
- State Key Laboratory of Bioelectronics, Southeast University, Nanjing, 210096, People's Republic of China
| |
Collapse
|
26
|
Chen H, Wu X, Zhou H, He Z, Li H, Wang Q. Epidermal growth factor upregulates the expression of A20 in hepatic cells via the MEK1/MSK1/p-p65 (Ser276) signaling pathway. Am J Transl Res 2021; 13:708-718. [PMID: 33594320 PMCID: PMC7868826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 11/04/2020] [Indexed: 06/12/2023]
Abstract
Tumor necrosis factor α-induced protein 3 (A20) suppresses inflammation by inhibiting the activation of nuclear factor kappa B (NF-κB). The aberrant expression of A20 is reportedly correlated with tumor development in human malignancies, including hepatocellular carcinoma (HCC). Proinflammatory mediators, including tumor necrosis factor α (TNF-α), interleukin-1, and lipopolysaccharide, may induce A20 expression. The present study revealed that epidermal growth factor (EGF) significantly increased A20 mRNA and protein levels in normal hepatic and hepatoma cells via the mitogen-activated protein kinase kinase-1 (MEK1)/mitogen- and stress-activated protein kinase-1 (MSK1)/phosphorylated (p)-p65 (Ser276) signaling pathway. A significant positive correlation was observed between the expression of EGF receptor and A20 in HCC and normal healthy liver tissues. The EGF-induced A20 upregulation was NF-κB-dependent and abolished by either the overexpression of the nuclear factor of a κ light polypeptide gene enhancer in a B-cell inhibitor α or treatment with the NF-κB inhibitor BAY11-7082. However, unlike TNF-α, EGF expression did not result in the upregulation of inflammatory molecules, including intercellular adhesion molecule 1, vascular cell adhesion molecule 1, and monocyte chemoattractant protein-1. These results indicate that EGF preferentially upregulated the protective mediator A20 over proinflammatory factors. To our knowledge, the present study is the first to demonstrate that EGF induced A20 expression by activating the MEK1/MSK1/p-p65 (Ser276) signaling pathway without causing an apparent inflammatory response. These results may further extend our understanding of liver inflammation and tumor development.
Collapse
Affiliation(s)
- Haiyang Chen
- Department of Internal Medicine, Henan Cancer Hospital, Affiliated Cancer Hospital of Zhengzhou UniversityZhengzhou 450008, Henan, P. R. China
| | - Xuan Wu
- Department of Internal Medicine, Henan Cancer Hospital, Affiliated Cancer Hospital of Zhengzhou UniversityZhengzhou 450008, Henan, P. R. China
| | - Hanqiong Zhou
- Department of Internal Medicine, Henan Cancer Hospital, Affiliated Cancer Hospital of Zhengzhou UniversityZhengzhou 450008, Henan, P. R. China
| | - Zhen He
- Department of Internal Medicine, Henan Cancer Hospital, Affiliated Cancer Hospital of Zhengzhou UniversityZhengzhou 450008, Henan, P. R. China
| | - Hongle Li
- Molecular Pathology Department, Affiliated Oncology Hospital, Zhengzhou UniversityZhengzhou 450008, Henan, P. R. China
| | - Qiming Wang
- Department of Internal Medicine, Henan Cancer Hospital, Affiliated Cancer Hospital of Zhengzhou UniversityZhengzhou 450008, Henan, P. R. China
| |
Collapse
|
27
|
Li J, Li X, Yuan Y, Wang Q, Xie L, Dai Y, Wang W, Li L, Lu X, Fan Q, Huang W. Efficient Polysulfide-Based Nanotheranostics for Triple-Negative Breast Cancer: Ratiometric Photoacoustics Monitored Tumor Microenvironment-Initiated H 2 S Therapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2020; 16:e2002939. [PMID: 32875678 DOI: 10.1002/smll.202002939] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Revised: 06/15/2020] [Indexed: 06/11/2023]
Abstract
The incidence of triple-negative breast cancer (TNBC) is difficult to predict, and TNBC has a high mortality rate among women worldwide. In this study, a theranostics approach is developed for TNBC with ratiometric photoacoustic monitored thiol-initiated hydrogen sulfide (H2 S) therapy. The ratiometric photoacoustic (PA) probe (CY) with a thiol-initiated H2 S donor (PSD) to form a nanosystem (CY-PSD nanoparticles) is integrated. In this theranostics approach, H2 S generated from PSD is sensed by CY based on ratiometric PA signals, which simultaneously pinpoints the tumor region. Additionally, H2 S is cytotoxic toward TNBC cells (MDA-MB 231), showing a tumor inhibition rate of 63%. To further verify its pharmacological mechanism, proteomics analysis is performed on tumors treated with CY-PSD nanoparticles. Cells are killed by the significant mitochondrial dysfunction via supressed energy supply and apoptosis initiation. Besides, the observed inhibition of oxidative stress also generates the cytotoxicity. Significant Kyoto Encyclopedia of Genes Genomes pathways related to TNBC are found to be inhibited. This H2 S theranostics approach updates the current anticancer therapies which brings promise for women suffering malignant breast cancer.
Collapse
Affiliation(s)
- Jie Li
- Jiangsu-Singapore Joint Research Center for Organic/Bio-Electronics & Information Displays and Institute of Advanced Materials (IAM), Nanjing Tech University, Nanjing, 211816, China
- Key Laboratory for Organic Electronics and Information Displays & Institute of Advanced Materials (IAM), Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM), Nanjing University of Posts & Telecommunications, Nanjing, 210023, China
| | - Xiang Li
- Key Laboratory for Organic Electronics and Information Displays & Institute of Advanced Materials (IAM), Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM), Nanjing University of Posts & Telecommunications, Nanjing, 210023, China
| | - Yan Yuan
- Jiangsu-Singapore Joint Research Center for Organic/Bio-Electronics & Information Displays and Institute of Advanced Materials (IAM), Nanjing Tech University, Nanjing, 211816, China
| | - Qi Wang
- Key Laboratory for Organic Electronics and Information Displays & Institute of Advanced Materials (IAM), Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM), Nanjing University of Posts & Telecommunications, Nanjing, 210023, China
| | - Lisi Xie
- Cancer Centre and Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macau, Macau SAR, 999078, China
| | - Yunlu Dai
- Cancer Centre and Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macau, Macau SAR, 999078, China
| | - Wenjun Wang
- Key Lab of Optical Communication Science and Technology of Shandong Province & School of Physics Science and Information Engineering, Liaocheng University, Liaocheng, 252059, China
| | - Lin Li
- Jiangsu-Singapore Joint Research Center for Organic/Bio-Electronics & Information Displays and Institute of Advanced Materials (IAM), Nanjing Tech University, Nanjing, 211816, China
| | - Xiaomei Lu
- Jiangsu-Singapore Joint Research Center for Organic/Bio-Electronics & Information Displays and Institute of Advanced Materials (IAM), Nanjing Tech University, Nanjing, 211816, China
| | - Quli Fan
- Key Laboratory for Organic Electronics and Information Displays & Institute of Advanced Materials (IAM), Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM), Nanjing University of Posts & Telecommunications, Nanjing, 210023, China
| | - Wei Huang
- Jiangsu-Singapore Joint Research Center for Organic/Bio-Electronics & Information Displays and Institute of Advanced Materials (IAM), Nanjing Tech University, Nanjing, 211816, China
| |
Collapse
|
28
|
Abdoli Shadbad M, Hajiasgharzadeh K, Baradaran B. Cross-talk between myeloid-derived suppressor cells and Mucin1 in breast cancer vaccination: On the verge of a breakthrough. Life Sci 2020; 258:118128. [PMID: 32710947 DOI: 10.1016/j.lfs.2020.118128] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Revised: 07/16/2020] [Accepted: 07/17/2020] [Indexed: 01/22/2023]
Abstract
Although breast cancer is one of the leading troublesome cancers, the available therapeutic options have not fulfilled the desired outcomes. Immune-based therapy has gained special attention for breast cancer treatment. Although this approach is highly tolerable, its low response rate has rendered it as an undesirable approach. This review aims to describe the essential oncogenic pathways involved in breast cancer, elucidate the immunosuppression and oncogenic effect of Mucin1, and introduce myeloid-derived suppressor cells, which are the main culprits of anti-tumoral immune response attenuation. The various auto-inductive loops between Mucin1 and myeloid-derived suppressor cells are focal in the suppression of anti-tumoral immune responses in patients with breast cancer. These cross-talks between the Mucin1 and myeloid-derived suppressor cells can be the underlying causes of immunotherapy's impotence for patients with breast cancer. This approach can pave the road for the development of a potent vaccine for patients with breast cancer and is translated into clinical settings.
Collapse
Affiliation(s)
| | - Khalil Hajiasgharzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Connective Tissue Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Pharmaceutical Analysis Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Neurosciences Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
29
|
Tang D, Ma J, Chu Z, Wang X, Zhao W, Zhang Q. Apatinib-induced NF-κB inactivation sensitizes triple-negative breast cancer cells to doxorubicin. Am J Transl Res 2020; 12:3741-3753. [PMID: 32774731 PMCID: PMC7407711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Accepted: 06/17/2020] [Indexed: 06/11/2023]
Abstract
Resistance to doxorubicin (DOX) is a major clinical challenge in triple-negative breast cancer (TNBC), which is highly diverse in different patients with variable outcomes. Apatinib is a new antiangiogenic agent, which has been reported to induce apoptosis. Nevertheless, the potential role and underlying mechanisms of apatinib in reversing DOX resistance of TNBC remain unknown. This work aims to evaluate the effects of apatinib on improving the sensitivity of TNBC cells to DOX and its underlying molecular basis. Our data indicate that apatinib treatment sensitizes DOX-resistant breast cancer cells to DOX, which is accompanied by significantly increased apoptosis. Additionally, this increased induction of apoptosis is associated with an enhancement of reactive oxygen species (ROS) accumulation. Importantly, it was found that followed by DOX treatment, apatinib could inhibit NF-κB signaling pathways, which have been validated to increase ROS production and reverse DOX resistance. Moreover, our in vivo results indicate the combination of DOX and apatinib exerted increased antitumor effects on TNBC cell xenograft models. Taken together, our study suggests that apatinib sensitizes TNBC cells to DOX in vitro and in vivo through inactivation of NF-κB signaling pathways, providing a rationale for the combined use of apatinib and DOX in TNBC chemotherapy.
Collapse
Affiliation(s)
- Dabei Tang
- Department of Medical Oncology, Harbin Medical University Cancer HospitalHaping Road No. 150, Harbin, Heilongjiang Province, China
| | - Jianli Ma
- Department of Radiotherapy, Harbin Medical University Cancer HospitalHaping Road No. 150, Harbin, Heilongjiang Province, China
| | - Zhong Chu
- Department of Medical Oncology, Harbin Medical University Cancer HospitalHaping Road No. 150, Harbin, Heilongjiang Province, China
| | - Xiaowei Wang
- Department of Medical Oncology, Harbin Medical University Cancer HospitalHaping Road No. 150, Harbin, Heilongjiang Province, China
| | - Wenhui Zhao
- Department of Medical Oncology, Harbin Medical University Cancer HospitalHaping Road No. 150, Harbin, Heilongjiang Province, China
| | - Qingyuan Zhang
- Department of Medical Oncology, Harbin Medical University Cancer HospitalHaping Road No. 150, Harbin, Heilongjiang Province, China
| |
Collapse
|
30
|
Thiebaut C, Konan HP, Guerquin MJ, Chesnel A, Livera G, Le Romancer M, Dumond H. The Role of ERα36 in Development and Tumor Malignancy. Int J Mol Sci 2020; 21:E4116. [PMID: 32526980 PMCID: PMC7312586 DOI: 10.3390/ijms21114116] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 06/03/2020] [Accepted: 06/04/2020] [Indexed: 02/06/2023] Open
Abstract
Estrogen nuclear receptors, represented by the canonical forms ERα66 and ERβ1, are the main mediators of the estrogen-dependent pathophysiology in mammals. However, numerous isoforms have been identified, stimulating unconventional estrogen response pathways leading to complex cellular and tissue responses. The estrogen receptor variant, ERα36, was cloned in 2005 and is mainly described in the literature to be involved in the progression of mammary tumors and in the acquired resistance to anti-estrogen drugs, such as tamoxifen. In this review, we will first specify the place that ERα36 currently occupies within the diversity of nuclear and membrane estrogen receptors. We will then report recent data on the impact of ERα36 expression and/or activity in normal breast and testicular cells, but also in different types of tumors including mammary tumors, highlighting why ERα36 can now be considered as a marker of malignancy. Finally, we will explain how studying the regulation of ERα36 expression could provide new clues to counteract resistance to cancer treatments in hormone-sensitive tumors.
Collapse
Affiliation(s)
- Charlène Thiebaut
- Université de Lorraine, CNRS, CRAN, F-54000 Nancy, France; (C.T.); (A.C.)
| | - Henri-Philippe Konan
- Université de Lyon, F-69000 Lyon, France; (H.-P.K.); (M.L.R.)
- INSERM U1052, Centre de Recherche en Cancérologie de Lyon, F-69000 Lyon, France
- CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, F-69000 Lyon, France
| | - Marie-Justine Guerquin
- Laboratory of Development of the Gonads, UMRE008 Genetic Stability Stem Cells and Radiation, Université de Paris, Université Paris Saclay, CEA, F-92265 Fontenay aux Roses, France; (M.-J.G.); (G.L.)
| | - Amand Chesnel
- Université de Lorraine, CNRS, CRAN, F-54000 Nancy, France; (C.T.); (A.C.)
| | - Gabriel Livera
- Laboratory of Development of the Gonads, UMRE008 Genetic Stability Stem Cells and Radiation, Université de Paris, Université Paris Saclay, CEA, F-92265 Fontenay aux Roses, France; (M.-J.G.); (G.L.)
| | - Muriel Le Romancer
- Université de Lyon, F-69000 Lyon, France; (H.-P.K.); (M.L.R.)
- INSERM U1052, Centre de Recherche en Cancérologie de Lyon, F-69000 Lyon, France
- CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, F-69000 Lyon, France
| | - Hélène Dumond
- Université de Lorraine, CNRS, CRAN, F-54000 Nancy, France; (C.T.); (A.C.)
| |
Collapse
|
31
|
Reddy TP, Choi DS, Anselme AC, Qian W, Chen W, Lantto J, Horak ID, Kragh M, Chang JC, Rosato RR. Simultaneous targeting of HER family pro-survival signaling with Pan-HER antibody mixture is highly effective in TNBC: a preclinical trial with PDXs. Breast Cancer Res 2020; 22:48. [PMID: 32414394 PMCID: PMC7227035 DOI: 10.1186/s13058-020-01280-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Accepted: 04/15/2020] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND The human epidermal growth factor receptor (HER) family, notably EGFR, is overexpressed in most triple-negative breast cancer (TNBC) cases and provides cancer cells with compensatory signals that greatly contribute to the survival and development of resistance in response to therapy. This study investigated the effects of Pan-HER (Symphogen, Ballerup, Denmark), a novel mixture of six monoclonal antibodies directed against members of the HER family EGFR, HER2, and HER3, in a preclinical trial of TNBC patient-derived xenografts (PDXs). METHODS Fifteen low passage TNBC PDX tumor samples were transferred into the right mammary fat pad of mice for engraftment. When tumors reached an average size of 100-200 mm3, mice were randomized (n ≥ 6 per group) and treated following three 1-week cycles consisting of three times/week intraperitoneal (IP) injection of either formulation buffer (vehicle control) or Pan-HER (50 mg/kg). At the end of treatment, tumors were collected for Western blot, RNA, and immunohistochemistry analyses. RESULTS All 15 TNBC PDXs were responsive to Pan-HER treatment, showing significant reductions in tumor growth consistent with Pan-HER-mediated tumor downmodulation of EGFR and HER3 protein levels and significantly decreased activation of associated HER family signaling pathways AKT and ERK. Tumor regression was observed in five of the models, which corresponded to those PDX tumor models with the highest level of HER family activation. CONCLUSIONS The marked effect of Pan-HER in numerous HER family-dependent TNBC PDX models justifies further studies of Pan-HER in TNBC clinical trials as a potential therapeutic option.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/pharmacology
- Cell Proliferation/drug effects
- Disease Models, Animal
- Drug Resistance, Neoplasm
- ErbB Receptors/antagonists & inhibitors
- ErbB Receptors/genetics
- ErbB Receptors/metabolism
- Female
- Humans
- Mice
- Molecular Targeted Therapy
- Mutation
- Receptor, ErbB-2/antagonists & inhibitors
- Receptor, ErbB-2/genetics
- Receptor, ErbB-2/metabolism
- Receptor, ErbB-3/antagonists & inhibitors
- Receptor, ErbB-3/genetics
- Receptor, ErbB-3/metabolism
- Triple Negative Breast Neoplasms/drug therapy
- Triple Negative Breast Neoplasms/genetics
- Triple Negative Breast Neoplasms/metabolism
- Triple Negative Breast Neoplasms/pathology
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- Tejaswini P Reddy
- Houston Methodist Cancer Center, Houston Methodist Hospital, Houston, TX, 77030, USA
- Texas A&M Health Science Center College of Medicine, Bryan, TX, 77807, USA
| | - Dong S Choi
- Houston Methodist Cancer Center, Houston Methodist Hospital, Houston, TX, 77030, USA
| | - Ann C Anselme
- Houston Methodist Cancer Center, Houston Methodist Hospital, Houston, TX, 77030, USA
- Texas A&M Health Science Center College of Medicine, Bryan, TX, 77807, USA
| | - Wei Qian
- Houston Methodist Cancer Center, Houston Methodist Hospital, Houston, TX, 77030, USA
| | - Wen Chen
- Houston Methodist Cancer Center, Houston Methodist Hospital, Houston, TX, 77030, USA
| | - Johan Lantto
- Symphogen A/S, Pederstrupvej 93, DK-2750, Ballerup, Denmark
| | - Ivan D Horak
- Symphogen A/S, Pederstrupvej 93, DK-2750, Ballerup, Denmark
| | - Michael Kragh
- Texas A&M Health Science Center College of Medicine, Bryan, TX, 77807, USA
| | - Jenny C Chang
- Houston Methodist Cancer Center, Houston Methodist Hospital, Houston, TX, 77030, USA
| | - Roberto R Rosato
- Houston Methodist Cancer Center, Houston Methodist Hospital, Houston, TX, 77030, USA.
| |
Collapse
|
32
|
Li H, Xu F, Gao G, Gao X, Wu B, Zheng C, Wang P, Li Z, Hua H, Li D. Hydrogen sulfide and its donors: Novel antitumor and antimetastatic therapies for triple-negative breast cancer. Redox Biol 2020; 34:101564. [PMID: 32403079 PMCID: PMC7218030 DOI: 10.1016/j.redox.2020.101564] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 04/27/2020] [Accepted: 04/28/2020] [Indexed: 12/20/2022] Open
Abstract
Hydrogen sulfide (H2S) is considered as a novel second-messenger molecule associated with the modulation of various physiological and pathological processes. In the field of antitumor research, endogenous H2S induces angiogenesis, accelerates the cell cycle and inhibits apoptosis, which results in promoting oncogenesis eventually. Interestingly, high concentrations of exogenous H2S liberated from donors suppress the growth of various tumors via inducing cellular acidification and modulating several signaling pathways involved in cell cycle regulation, proliferation, apoptosis and metastasis. The selective release of certain concentrations of H2S from H2S donors in the target has been considered as an alternative tumor therapy strategy. Triple-negative breast cancer (TNBC), an aggressive subtype with less than one year median survival time, is known to account for approximately 15–20% of all breast cancers. Due to the lack of approved targeted therapy, the clinical treatment of TNBC is still hindered by metastasis as well as recurrence. Significant efforts have been spent on developing novel treatments of TNBC, and remarkable progress in the control of TNBC by H2S donors and their derivatives have been made in recent years. This review summarizes various pathways involved in antitumor and anti-metastasis effects of H2S donors and their derivatives on TNBC, which provides novel insights for TNBC treatment.
Collapse
Affiliation(s)
- Haonan Li
- Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, 110016, PR China
| | - Fanxing Xu
- Wuya College of Innovation, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, 110016, PR China
| | - Gang Gao
- Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, 110016, PR China
| | - Xiang Gao
- Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, 110016, PR China
| | - Bo Wu
- Molecular Imaging Laboratory, MGH/MIT/HMS Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital/Harvard Medical School, Building 75, Charlestown, MA, 02129, United States
| | - Chao Zheng
- PET Center, Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, CT, 06520, United States
| | - Peng Wang
- Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, Nanjing, 210009, PR China
| | - Zhanlin Li
- Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, 110016, PR China
| | - Huiming Hua
- Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, 110016, PR China
| | - Dahong Li
- Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, 110016, PR China.
| |
Collapse
|
33
|
Fan Z, Kim S, Bai Y, Diergaarde B, Park HJ. 3'-UTR Shortening Contributes to Subtype-Specific Cancer Growth by Breaking Stable ceRNA Crosstalk of Housekeeping Genes. Front Bioeng Biotechnol 2020; 8:334. [PMID: 32411683 PMCID: PMC7201092 DOI: 10.3389/fbioe.2020.00334] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Accepted: 03/25/2020] [Indexed: 12/21/2022] Open
Abstract
Shortening of 3'UTRs (3'US) through alternative polyadenylation is a post-transcriptional mechanism that regulates the expression of hundreds of genes in human cancers. In breast cancer, different subtypes of tumor samples, such as estrogen receptor positive and negative (ER+ and ER-), are characterized by distinct molecular mechanisms, suggesting possible differences in the post-transcriptional regulation between the subtype tumors. In this study, based on the profound tumorigenic role of 3'US interacting with competing-endogenous RNA (ceRNA) network (3'US-ceRNA effect), we hypothesize that the 3'US-ceRNA effect drives subtype-specific tumor growth. However, we found that the subtypes are available in different sample sizes, biasing the ceRNA network size and disabling the fair comparison of the 3'US-ceRNA effect. Using normalized Laplacian matrix eigenvalue distribution, we addressed this bias and built tumor ceRNA networks comparable between the subtypes. Based on the comparison, we identified a novel role of housekeeping (HK) genes as stable and strong miRNA sponges (sponge HK genes) that synchronize the ceRNA networks of normal samples (adjacent to ER+ and ER- tumor samples). We further found that distinct 3'US events in the ER- tumor break the stable sponge effect of HK genes in a subtype-specific fashion, especially in association with the aggressive and metastatic phenotypes. Knockdown of NUDT21 further suggested the role of 3'US-ceRNA effect in repressing HK genes for tumor growth. In this study, we identified 3'US-ceRNA effect on the sponge HK genes for subtype-specific growth of ER- tumors.
Collapse
Affiliation(s)
- Zhenjiang Fan
- Department of Computer Science, University of Pittsburgh, Pittsburgh, PA, United States
| | - Soyeon Kim
- Department of Pediatrics, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
- Division of Pulmonary Medicine, Children's Hospital of Pittsburgh UPMC, Pittsburgh, PA, United States
| | - Yulong Bai
- Department of Human Genetics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, United States
| | - Brenda Diergaarde
- Department of Human Genetics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, United States
- Hillman Cancer Center, University of Pittsburgh Medical Cancer, Pittsburgh, PA, United States
| | - Hyun Jung Park
- Department of Human Genetics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, United States
| |
Collapse
|
34
|
Liu W, Cheng L, Li Q, Jing J. TRIP6 regulates the proliferation, migration, invasion and apoptosis of osteosarcoma cells by activating the NF-κB signaling pathway. Exp Ther Med 2020; 19:2317-2325. [PMID: 32104300 PMCID: PMC7027267 DOI: 10.3892/etm.2020.8466] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Accepted: 12/18/2019] [Indexed: 11/06/2022] Open
Abstract
Thyroid hormone receptor-interacting protein 6 (TRIP6), a member of the zyxin family of Lin-Isl-Mec (LIM) proteins, is an adaptor protein primarily expressed in epithelial cells. TRIP6 can regulate a variety of cellular responses, such as actin cytoskeletal reorganization and cell adhesion. However, to the best of our knowledge, the role of TRIP6 in osteosarcoma (Os) has not been previously reported. Therefore, the present study investigated the role of TRIP6 in the occurrence and development of Os, and the potential of utilizing TRIP6 as a therapeutic target in Os. The present results suggested that the expression levels of TRIP6 were significantly increased in Os cells and clinical tissue specimens compared with normal osteoblasts and adjacent non-tumor tissue. Moreover, the present results suggested that overexpressing TRIP6 significantly increased proliferation, migration and invasion, while inhibiting apoptosis in Os cells. However, silencing TRIP6 decreased proliferation, migration and invasion, while activating apoptosis in Os cells. The present results suggested that overexpression of TRIP6 increased NF-κB activation by decreasing the protein expression levels of inhibitor of κBα, and increasing total and phosphorylated P65 levels. The present results indicated that TRIP6 silencing decreased NF-κB activation. Collectively, the present results suggested that TRIP6 may play a role in promoting Os cell proliferation, migration and invasion, while inhibiting cell apoptosis. Furthermore, TRIP6 may be utilized as a novel prognostic biomarker and therapeutic target in Os.
Collapse
Affiliation(s)
- Wei Liu
- Department of Orthopedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230601, P.R. China
| | - Li Cheng
- Department of Orthopedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230601, P.R. China
| | - Qingning Li
- Department of Orthopedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230601, P.R. China
| | - Juehua Jing
- Department of Orthopedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230601, P.R. China
| |
Collapse
|
35
|
Dai W, Wu J, Wang D, Wang J. Cancer gene therapy by NF-κB-activated cancer cell-specific expression of CRISPR/Cas9 targeting telomeres. Gene Ther 2020; 27:266-280. [DOI: 10.1038/s41434-020-0128-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Revised: 01/22/2020] [Accepted: 01/28/2020] [Indexed: 02/07/2023]
|
36
|
Kaempferia parviflora Extract Inhibits STAT3 Activation and Interleukin-6 Production in HeLa Cervical Cancer Cells. Int J Mol Sci 2019; 20:ijms20174226. [PMID: 31470515 PMCID: PMC6747281 DOI: 10.3390/ijms20174226] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 08/23/2019] [Accepted: 08/27/2019] [Indexed: 02/06/2023] Open
Abstract
Kaempferia parviflora (KP) has been reported to have anti-cancer activities. We previously reported its effects against cervical cancer cells and continued to elucidate the effects of KP on inhibiting the production and secretion of interleukin (IL)-6, as well as its relevant signaling pathways involved in cervical tumorigenesis. We discovered that KP suppressed epidermal growth factor (EGF)-induced IL-6 secretion in HeLa cells, and it was associated with a reduced level of Glycoprotein 130 (GP130), phosphorylated signal transducers and activators of transcription 3 (STAT3), and Mcl-1. Our data clearly showed that KP has no effect on nuclear factor kappa B (NF-κB) localization status. However, we found that KP inhibited EGF-stimulated phosphorylation of tyrosine 1045 and tyrosine 1068 of EGF receptor (EGFR) without affecting its expression level. The inhibition of EGFR activation was verified by the observation that KP significantly suppressed a major downstream MAP kinase, ERK1/2. Consistently, KP reduced the expression of Ki-67 protein, which is a cellular marker for proliferation. Moreover, KP potently inhibited phosphorylation of STAT3, Akt, and the expression of Mcl-1 in response to exogenous IL-6 stimulation. These data suggest that KP suppresses EGF-induced production of IL-6 and inhibits its autocrine IL-6/STAT3 signaling critical for maintaining cancer cell progression. We believe that KP may be a potential alternative anti-cancer agent for suppressing cervical tumorigenesis.
Collapse
|
37
|
Shi F, Deng Z, Zhou Z, Jiang B, Jiang CY, Zhao RZ, Sun F, Cui D, Sun MH, Sun Q, Wang XJ, Wu Q, Xia SJ, Han BM. Heat injured stromal cells-derived exosomal EGFR enhances prostatic wound healing after thulium laser resection through EMT and NF-κB signaling. Prostate 2019; 79:1238-1255. [PMID: 31124594 DOI: 10.1002/pros.23827] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2019] [Accepted: 04/23/2019] [Indexed: 12/25/2022]
Abstract
BACKGROUND This study investigated shallow heat injury to prostate stromal fibroblasts and epithelial cells and their interaction to regulate the wound healing and the underlying molecular events. METHODS Prostate stromal fibroblasts and epithelial cells were cultured individually or cocultured and subjected to shallow heat injury for assessments of cell proliferation, migration, apoptosis, cell cycle distribution, and gene expression. The supernatant of heat-injured WPMY-1 cells was collected for exosome extraction and assessments. Furthermore, beagle dogs received thulium laser resection of the prostate (TmLRP) and randomly divided into Gefitinib, GW4869, and control treatment for the histological analysis, tissue re-epithelialization, and epidermal growth factor receptor (EGFR) expression on the prostatic wound surface. Immunofluorescence was to evaluate p63-positive basal progenitor cell trans-differentiation and macrophage polarization and ELISA was to detect cytokine levels in beagles' urine. RESULTS Shallow heat injury caused these cells to enter a stressed state and enhanced their crosstalk. The prostate stromal fibroblasts produced and secreted more exosomal-EGFR and other cytokines and chemokines after shallow heat injury, resulting in increased proliferation and migration of prostate epithelial cells during wound healing. The wound healing of the canine prostatic urethra following the TmLRP procedure was slower in the Gefitinib and GW4869 treatment group than in the control group of animals. Immunofluorescence and ELISA showed that reduced EGFR expression interrupted macrophage polarization but increased the inflammatory response. CONCLUSIONS Shallow heat injury was able to promote the interaction of prostate stromal cells with prostate epithelial cells to enhance wound healing. Stromal-derived exosomal-EGFR plays a crucial role in the balance of the macrophage polarization and prostatic wound healing.
Collapse
Affiliation(s)
- Fei Shi
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Zheng Deng
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Zheng Zhou
- Department of Urology, Shanghai General Hospital Affiliated to Nanjing Medical University, Shanghai, China
| | - Bo Jiang
- Department of Urology, Qingdao Municipal Hospital, School of Medicine, Qingdao University, Qingdao, Shandong, China
| | - Chen-Yi Jiang
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Rui-Zhe Zhao
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Feng Sun
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
- Institute of Urology, Shanghai Jiao Tong University, Shanghai, China
| | - Di Cui
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
- Institute of Urology, Shanghai Jiao Tong University, Shanghai, China
| | - Meng-Hao Sun
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
- Institute of Urology, Shanghai Jiao Tong University, Shanghai, China
| | - Qian Sun
- Department of Urology, Shanghai General Hospital Affiliated to Nanjing Medical University, Shanghai, China
| | - Xing-Jie Wang
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
- Institute of Urology, Shanghai Jiao Tong University, Shanghai, China
| | - Qi Wu
- Department of Urology, Shanghai General Hospital Affiliated to Nanjing Medical University, Shanghai, China
| | - Shu-Jie Xia
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
- Institute of Urology, Shanghai Jiao Tong University, Shanghai, China
| | - Bang-Min Han
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
- Institute of Urology, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
38
|
Prajoko YW, Aryandono T. The Effect of P-Glycoprotein (P-gp), Nuclear Factor-Kappa B (Nf-κb), and Aldehyde Dehydrogenase-1 (ALDH-1) Expression on Metastases, Recurrence and Survival in Advanced Breast Cancer Patients. Asian Pac J Cancer Prev 2019; 20:1511-1518. [PMID: 31128056 PMCID: PMC6857879 DOI: 10.31557/apjcp.2019.20.5.1511] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Objective: To investigate the level of three drug resistance proteins; P-glycoprotein 1 (P-gp), nuclear factor
kappa-light-chain-enhancer of activated B cells (NF-κB) and aldehyde dehydrogenase isoform 1 (ALDH1) expression
and their relationship to metastasis, recurrence and survival in advanced breast cancer patients that received neoadjuvant
chemotherapy. Methods: This study is a combination of prospective and retrospective cohort study involving one
hundred and thirty one cases of advanced stage invasive breast cancer that have received neoadjuvant chemotherapy.
Initial biopsy specimens (incisional biopsy or core biopsy) were taken from paraffin blocks. Immunohistochemistry
(IHC) was used to detect P-gp, NF-κB, and ALDH1 expression. Prospectively analysed patients were followed for five
years and evaluated for recurrence and death. Results: The expression of P-gp has no significant statistical correlation
to metastases (p = 0.659), recurrence (p = 0.862) and survival (p = 0.835) in advanced stage breast cancer patients
who received neoadjuvant chemotherapy. Similarly, ALDH1 was not correlated to metastases (p=0.120), recurrence
(p = 0.186) and survival (p = 0.254) statistically. We found that NF-κB expression showed a significant correlation to
metastases (p=0.004), recurrence (p = 0.016) and overall survival (p = 0.041) in advanced stage breast cancer patients
after neoadjuvant chemotherapy. Conclusion: NF-κB expression is a potential marker that can be used to assess or
to predict increasing risk of metastases, recurrence and survival in advanced stage breast cancer patients who receive
neoadjuvant chemotherapy.
Collapse
Affiliation(s)
- Yan Wisnu Prajoko
- Department of Oncologic Surgery, Faculty of Medicine, Universitas Diponegoro, Semarang, Indonesia.
| | - Teguh Aryandono
- Department of Oncologic Surgery, Faculty of Medicine, Universitas Diponegoro, Semarang, Indonesia.
| |
Collapse
|
39
|
Wang D, Dai W, Wang J. A Cell-Specific Nuclear Factor-Kappa B–Activating Gene Expression Strategy for Delivering Cancer Immunotherapy. Hum Gene Ther 2019; 30:471-484. [DOI: 10.1089/hum.2018.093] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Affiliation(s)
- Danyang Wang
- State Key Laboratory of Bioelectronics, Southeast University, Nanjing, P.R. China
| | - Wei Dai
- State Key Laboratory of Bioelectronics, Southeast University, Nanjing, P.R. China
| | - Jinke Wang
- State Key Laboratory of Bioelectronics, Southeast University, Nanjing, P.R. China
| |
Collapse
|
40
|
Zhang S, Lin X. CARMA3: Scaffold Protein Involved in NF-κB Signaling. Front Immunol 2019; 10:176. [PMID: 30814996 PMCID: PMC6381293 DOI: 10.3389/fimmu.2019.00176] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2018] [Accepted: 01/21/2019] [Indexed: 12/26/2022] Open
Abstract
Scaffold proteins are defined as pivotal molecules that connect upstream receptors to specific effector molecules. Caspase recruitment domain protein 10 (CARD10) gene encodes a scaffold protein CARMA3, belongs to the family of CARD and membrane-associated guanylate kinase-like protein (CARMA). During the past decade, investigating the function of CARMA3 has revealed that it forms a complex with BCL10 and MALT1 to mediate different receptors-dependent signaling, including GPCR and EGFR, leading to activation of the transcription factor NF-κB. More recently, CARMA3 and its partners are also reported to be involved in antiviral innate immune response and DNA damage response. In this review, we summarize the biology of CARMA3 in multiple receptor-induced NF-κB signaling. Especially, we focus on discussing the function of CARMA3 in regulating NF-κB activation and antiviral IFN signaling in the context of recent progress in the field.
Collapse
Affiliation(s)
| | - Xin Lin
- Department of Basic Medical Sciences, Tsinghua University School of Medicine, Beijing, China
| |
Collapse
|
41
|
Mussbacher M, Salzmann M, Brostjan C, Hoesel B, Schoergenhofer C, Datler H, Hohensinner P, Basílio J, Petzelbauer P, Assinger A, Schmid JA. Cell Type-Specific Roles of NF-κB Linking Inflammation and Thrombosis. Front Immunol 2019; 10:85. [PMID: 30778349 PMCID: PMC6369217 DOI: 10.3389/fimmu.2019.00085] [Citation(s) in RCA: 398] [Impact Index Per Article: 66.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Accepted: 01/11/2019] [Indexed: 12/22/2022] Open
Abstract
The transcription factor NF-κB is a central mediator of inflammation with multiple links to thrombotic processes. In this review, we focus on the role of NF-κB signaling in cell types within the vasculature and the circulation that are involved in thrombo-inflammatory processes. All these cells express NF-κB, which mediates important functions in cellular interactions, cell survival and differentiation, as well as expression of cytokines, chemokines, and coagulation factors. Even platelets, as anucleated cells, contain NF-κB family members and their corresponding signaling molecules, which are involved in platelet activation, as well as secondary feedback circuits. The response of endothelial cells to inflammation and NF-κB activation is characterized by the induction of adhesion molecules promoting binding and transmigration of leukocytes, while simultaneously increasing their thrombogenic potential. Paracrine signaling from endothelial cells activates NF-κB in vascular smooth muscle cells and causes a phenotypic switch to a “synthetic” state associated with a decrease in contractile proteins. Monocytes react to inflammatory situations with enforced expression of tissue factor and after differentiation to macrophages with altered polarization. Neutrophils respond with an extension of their life span—and upon full activation they can expel their DNA thereby forming so-called neutrophil extracellular traps (NETs), which exert antibacterial functions, but also induce a strong coagulatory response. This may cause formation of microthrombi that are important for the immobilization of pathogens, a process designated as immunothrombosis. However, deregulation of the complex cellular links between inflammation and thrombosis by unrestrained NET formation or the loss of the endothelial layer due to mechanical rupture or erosion can result in rapid activation and aggregation of platelets and the manifestation of thrombo-inflammatory diseases. Sepsis is an important example of such a disorder caused by a dysregulated host response to infection finally leading to severe coagulopathies. NF-κB is critically involved in these pathophysiological processes as it induces both inflammatory and thrombotic responses.
Collapse
Affiliation(s)
- Marion Mussbacher
- Institute of Vascular Biology and Thrombosis Research, Medical University of Vienna, Vienna, Austria
| | - Manuel Salzmann
- Institute of Vascular Biology and Thrombosis Research, Medical University of Vienna, Vienna, Austria
| | - Christine Brostjan
- Department of Surgery, General Hospital, Medical University of Vienna, Vienna, Austria
| | - Bastian Hoesel
- Institute of Vascular Biology and Thrombosis Research, Medical University of Vienna, Vienna, Austria
| | | | - Hannes Datler
- Institute of Vascular Biology and Thrombosis Research, Medical University of Vienna, Vienna, Austria
| | - Philipp Hohensinner
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
| | - José Basílio
- Institute of Vascular Biology and Thrombosis Research, Medical University of Vienna, Vienna, Austria
| | - Peter Petzelbauer
- Skin and Endothelial Research Division, Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Alice Assinger
- Institute of Vascular Biology and Thrombosis Research, Medical University of Vienna, Vienna, Austria
| | - Johannes A Schmid
- Institute of Vascular Biology and Thrombosis Research, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
42
|
Jin MZ, Xia BR, Xu Y, Jin WL. Curaxin CBL0137 Exerts Anticancer Activity via Diverse Mechanisms. Front Oncol 2018; 8:598. [PMID: 30581774 PMCID: PMC6292929 DOI: 10.3389/fonc.2018.00598] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Accepted: 11/26/2018] [Indexed: 12/22/2022] Open
Abstract
Chemotherapy with or without radiation remains the first choice for most cancers. However, intolerant side effects and conventional drug resistance restrict actual clinical efficacy. Curaxin CBL0137 is designed to regulate p53 and nuclear factor-κB simultaneously and to prevent the resistance caused by a single target. Functionally, CBL0137 exhibits an antitumor activity in multiple cancers, including glioblastoma, renal cell carcinoma, melanoma, neuroblastoma, and small cell lung cancer (SCLC). Mechanistically, CBL0137 is originally identified to act by facilitates chromatin transcription (FACT) complex. Further investigations reveal that several pathways, such as NOTCH1 and heat shock factor 1 (HSF1), are involved in the process. CBL0137 has been reported to target cancer stem cells (CSCs) and enhance chemotherapy/monotherapy efficacy. The translational advance of CBL0137 into clinical practice is expected to provide a promising future for cancer treatment.
Collapse
Affiliation(s)
- Ming-Zhu Jin
- Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bai-Rong Xia
- Department of Gynecology, The Affiliated Tumor Hospital, Harbin Medical University, Harbin, China
| | - Yu Xu
- Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wei-Lin Jin
- Key Laboratory for Thin Film and Microfabrication Technology of Ministry of Education, Department of Instrument Science and Engineering, Shanghai Engineering Center for Intelligent Diagnosis and Treatment Instrument, School of Electronic Information and Electronic Engineering, Institute of Nano Biomedicine and Engineering, Shanghai Jiao Tong University, Shanghai, China.,National Center for Translational Medicine, Collaborative Innovational Center for System Biology, Shanghai Jiao Tong University, Shanghai, China.,Shaanxi Key Laboratory of Brain Disorders and Institute of Basic and Translational Medicine, Xi'an Medical University, Xi'an, China
| |
Collapse
|
43
|
Park YMM, Steck SE, Fung TT, Merchant AT, Elizabeth Hodgson M, Keller JA, Sandler DP. Higher diet-dependent acid load is associated with risk of breast cancer: Findings from the sister study. Int J Cancer 2018; 144:1834-1843. [PMID: 30247761 DOI: 10.1002/ijc.31889] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Revised: 08/16/2018] [Accepted: 09/10/2018] [Indexed: 01/22/2023]
Abstract
Dietary factors that contribute to chronic low-grade metabolic acidosis have been linked to breast cancer risk, but to date no epidemiologic study has examined diet-dependent acid load and breast cancer. We used data from 43,570 Sister Study participants who completed a validated food frequency questionnaire at enrollment (2003-2009) and satisfied eligibility criteria. The Potential Renal Acid Load (PRAL) score was used to estimate diet-dependent acid load. Higher scores reflect greater consumption of protein and phosphorus, and lower consumption of potassium, calcium and magnesium. The association between PRAL and breast cancer was evaluated using multivariable Cox proportional hazards regression. We identified 1,614 invasive breast cancers diagnosed at least 1 year after enrollment (mean follow-up, 7.6 years). The highest PRAL quartile, reflecting greater acid-forming potential, was associated with increased risk of breast cancer (HRhighest vs. lowest quartile : 1.21 [95% CI, 1.04-1.41], ptrend = 0.04). The association was more pronounced for estrogen receptor (ER)-negative (HRhighest vs. lowest quartile : 1.67 [95% CI, 1.07-2.61], ptrend = 0.03) and triple-negative breast cancer (HRhighest vs. lowest quartile : 2.20 [95% CI, 1.23-3.95], ptrend = 0.02). Negative PRAL scores, representing consumption of alkaline diets, were associated with decreased risk of ER-negative and triple-negative breast cancer, compared to a PRAL score of 0 representing neutral pH. Higher diet-dependent acid load may be a risk factor for breast cancer while alkaline diets may be protective. Since PRAL scores are positively correlated with meat consumption and negatively correlated with fruit and vegetable intake, results also suggest that diets high in fruits and vegetables and low in meat may be protective against hormone receptor negative breast cancer.
Collapse
Affiliation(s)
- Yong-Moon Mark Park
- Epidemiology Branch, National Institute of Environmental Health Sciences, Research Triangle Park, NC
| | - Susan E Steck
- Cancer Prevention and Control Program, University of South Carolina, Columbia, SC.,Department of Epidemiology and Biostatistics, Arnold School of Public Health, University of South Carolina, Columbia, SC
| | - Teresa T Fung
- Department of Nutrition, Simmons College, Boston, MA.,Department of Nutrition, Harvard TH Chan School of Public Health, Boston, MA
| | - Anwar T Merchant
- Department of Epidemiology and Biostatistics, Arnold School of Public Health, University of South Carolina, Columbia, SC
| | | | | | - Dale P Sandler
- Epidemiology Branch, National Institute of Environmental Health Sciences, Research Triangle Park, NC
| |
Collapse
|
44
|
The flaxseed lignan secoisolariciresinol diglucoside decreases local inflammation, suppresses NFκB signaling, and inhibits mammary tumor growth. Breast Cancer Res Treat 2018; 173:545-557. [PMID: 30367332 PMCID: PMC6394576 DOI: 10.1007/s10549-018-5021-6] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Accepted: 10/20/2018] [Indexed: 12/31/2022]
Abstract
PURPOSE Exposure to the polyphenolic plant lignan secoisolariciresinol diglucoside (SDG) and its metabolite enterolactone (ENL) has been associated with reduced breast cancer progression, particularly for estrogen receptor alpha (ERα)-negative disease, and decreased preclinical mammary tumor growth. However, while preclinical studies have established that SDG and ENL affect measures of progression in models of triple-negative breast cancer (TNBC, a subset of ERα-negative disease), the molecular mechanisms underlying these effects remain unclear. METHODS C57BL/6 mice were fed a control diet (control, 10% kcal from fat) or control diet + SDG (SDG, 100 mg/kg diet) for 8 weeks, then orthotopically injected with syngeneic E0771 mammary tumor cells (a model of TNBC); tumor growth was monitored for 3 weeks. The role of reduced NF-κB signaling in SDG's anti-tumor effects was explored in vitro via treatment with the bioactive SDG metabolite ENL. In addition to the murine E0771 cells, the in vitro studies utilized MDA-MB-231 and MCF-7 cells, two human cell lines which model the triple-negative and luminal A breast cancer subtypes, respectively. RESULTS SDG supplementation in the mice significantly reduced tumor volume and expression of phospho-p65 and NF-κB target genes (P < 0.05). Markers of macrophage infiltration were decreased in the distal-to-tumor mammary fat pad of mice supplemented with SDG relative to control mice (P < 0.05). In vitro, ENL treatment inhibited viability, survival, and NF-κB activity and target gene expression in E0771, MDA-MB-231, and MCF-7 cells (P < 0.05). Overexpression of Rela attenuated ENL's inhibition of E0771 cell viability and survival. CONCLUSIONS SDG reduces tumor growth in the E0771 model of TNBC, likely via a mechanism involving inhibition of NF-κB activity. SDG could serve as a practical and effective adjuvant treatment to reduce recurrence, but greater understanding of its effects is needed to inform the development of more targeted recommendations for its use.
Collapse
|
45
|
Sierra JC, Asim M, Verriere TG, Piazuelo MB, Suarez G, Romero-Gallo J, Delgado AG, Wroblewski LE, Barry DP, Peek RM, Gobert AP, Wilson KT. Epidermal growth factor receptor inhibition downregulates Helicobacter pylori-induced epithelial inflammatory responses, DNA damage and gastric carcinogenesis. Gut 2018; 67:1247-1260. [PMID: 28473630 PMCID: PMC5671361 DOI: 10.1136/gutjnl-2016-312888] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Revised: 03/30/2017] [Accepted: 04/09/2017] [Indexed: 12/23/2022]
Abstract
OBJECTIVE Gastric cancer is the third leading cause of cancer death worldwide and infection by Helicobacter pylori is the strongest risk factor. We have reported increased epidermal growth factor receptor (EGFR) phosphorylation in the H. pylori-induced human carcinogenesis cascade, and association with DNA damage. Our goal was to determine the role of EGFR activation in gastric carcinogenesis. DESIGN We evaluated gefitinib, a specific EGFR inhibitor, in chemoprevention of H. pylori-induced gastric inflammation and cancer development. Mice with genetically targeted epithelial cell-specific deletion of Egfr (EfgrΔepi mice) were also used. RESULTS In C57BL/6 mice, gefitinib decreased Cxcl1 and Cxcl2 expression by gastric epithelial cells, myeloperoxidase-positive inflammatory cells in the mucosa and epithelial DNA damage induced by H. pylori infection. Similar reductions in chemokines, inflammatory cells and DNA damage occurred in infected EgfrΔepi versus Egfrfl/fl control mice. In H. pylori-infected transgenic insulin-gastrin (INS-GAS) mice and gerbils, gefitinib treatment markedly reduced dysplasia and carcinoma. Gefitinib blocked H. pylori-induced activation of mitogen-activated protein kinase 1/3 (MAPK1/3) and activator protein 1 in gastric epithelial cells, resulting in inhibition of chemokine synthesis. MAPK1/3 phosphorylation and JUN activation was reduced in gastric tissues from infected wild-type and INS-GAS mice treated with gefitinib and in primary epithelial cells from EfgrΔepi versus Egfrfl/fl mice. Epithelial EGFR activation persisted in humans and mice after H. pylori eradication, and gefitinib reduced gastric carcinoma in INS-GAS mice treated with antibiotics. CONCLUSIONS These findings suggest that epithelial EGFR inhibition represents a potential strategy to prevent development of gastric carcinoma in H. pylori-infected individuals.
Collapse
Affiliation(s)
- Johanna C. Sierra
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Nashville, Tennessee, USA
| | - Mohammad Asim
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Nashville, Tennessee, USA
| | - Thomas G. Verriere
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Nashville, Tennessee, USA
| | - M. Blanca Piazuelo
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Nashville, Tennessee, USA
| | - Giovanni Suarez
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Nashville, Tennessee, USA
| | - Judith Romero-Gallo
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Nashville, Tennessee, USA
| | - Alberto G. Delgado
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Nashville, Tennessee, USA
| | - Lydia E. Wroblewski
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Nashville, Tennessee, USA
| | - Daniel P. Barry
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Nashville, Tennessee, USA
| | - Richard M. Peek
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Nashville, Tennessee, USA,Department of Cancer Biology, Nashville, Tennessee, USA,Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Alain P. Gobert
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Nashville, Tennessee, USA,Center for Mucosal Inflammation and Cancer, Nashville, Tennessee, USA
| | - Keith T. Wilson
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Nashville, Tennessee, USA,Department of Cancer Biology, Nashville, Tennessee, USA,Center for Mucosal Inflammation and Cancer, Nashville, Tennessee, USA,Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA,Veterans Affairs Tennessee Valley Healthcare System, Nashville, Tennessee, USA
| |
Collapse
|
46
|
Hollingsworth J, Lau A, Tone A, Kollara A, Allen L, Colgan TJ, Dube V, Rosen B, Murphy KJ, Greenblatt EM, Feigenberg T, Virtanen C, Brown TJ. BRCA1 Mutation Status and Follicular Fluid Exposure Alters NFκB Signaling and ISGylation in Human Fallopian Tube Epithelial Cells. Neoplasia 2018; 20:697-709. [PMID: 29852322 PMCID: PMC6030391 DOI: 10.1016/j.neo.2018.05.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Revised: 05/07/2018] [Accepted: 05/11/2018] [Indexed: 12/31/2022] Open
Abstract
Germline BRCA1 or BRCA2 mutations (mtBRCA1 and mtBRCA2) increase risk for high-grade serous ovarian cancer (HGSOC), the most commonly diagnosed epithelial ovarian cancer histotype. Other identified risk factors for this cancer, which originates primarily in the distal fallopian tube epithelium (FTE), implicate ovulation, during which the FTE cells become transiently exposed to follicular fluid (FF). To test whether mtBRCA1 or mtBRCA2 nonmalignant FTE cells respond differently to periovulatory FF exposure than control patient FTE cells, gene expression profiles from primary FTE cultures derived from BRCA1 or BRCA2 mutation carriers or control patients were compared at baseline, 24 hours after FF exposure, and 24 hours after FF replacement with culture medium. Hierarchical clustering revealed both FF exposure and BRCA mutation status affect gene expression, with BRCA1 mutation having the greatest impact. Gene set enrichment analysis revealed increased NFκB and EGFR signaling at baseline in mtBRCA1 samples, with increased interferon target gene expression, including members of the ISGylation pathway, observed after recovery from FF exposure. Gene set enrichment analysis did not identify altered pathway signaling in mtBRCA2 samples. An inverse relationship between EGFR signaling and ISGylation with BRCA1 protein levels was verified in an immortalized FTE cell line, OE-E6/E7, stably transfected with BRCA1 cDNA. Suppression of ISG15 and ISGylated protein levels by increased BRCA1 expression was found to be mediated by decreased NFκB signaling. These studies indicate that increased NFκB signaling associated with decreased BRCA1 expression results in increased ISG15 and protein ISGylation following FF exposure, which may be involved in predisposition to HGSOC.
Collapse
Affiliation(s)
- Julia Hollingsworth
- The Lunenfeld-Tanenbaum Research Institute of Mount Sinai Hospital, Toronto, ON; Institute of Medical Sciences, University of Toronto, Toronto, ON; Department of Obstetrics and Gynecology, University of Toronto, Toronto, ON
| | - Angela Lau
- The Lunenfeld-Tanenbaum Research Institute of Mount Sinai Hospital, Toronto, ON; Department of Obstetrics and Gynecology, University of Toronto, Toronto, ON; Department of Physiology, University of Toronto, Toronto, ON
| | - Alicia Tone
- Department of Obstetrics and Gynecology, University of Toronto, Toronto, ON; Division of Gynecologic Oncology, Princess Margaret Cancer Centre, Toronto, ON
| | - Alexandra Kollara
- The Lunenfeld-Tanenbaum Research Institute of Mount Sinai Hospital, Toronto, ON; Department of Obstetrics and Gynecology, University of Toronto, Toronto, ON
| | - Lisa Allen
- Department of Obstetrics and Gynecology, University of Toronto, Toronto, ON
| | - Terence J Colgan
- The Lunenfeld-Tanenbaum Research Institute of Mount Sinai Hospital, Toronto, ON; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON; Pathology and Laboratory Medicine, Mount Sinai Hospital, Toronto, ON
| | - Valerie Dube
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON; Department of Pathology, Women's College Hospital, Toronto, ON
| | - Barry Rosen
- Department of Obstetrics and Gynecology, University of Toronto, Toronto, ON; Division of Gynecologic Oncology, Princess Margaret Cancer Centre, Toronto, ON
| | - K Joan Murphy
- Department of Obstetrics and Gynecology, University of Toronto, Toronto, ON; Division of Gynecologic Oncology, Princess Margaret Cancer Centre, Toronto, ON
| | - Ellen M Greenblatt
- The Lunenfeld-Tanenbaum Research Institute of Mount Sinai Hospital, Toronto, ON; Department of Obstetrics and Gynecology, University of Toronto, Toronto, ON
| | - Tomer Feigenberg
- Department of Obstetrics and Gynecology, University of Toronto, Toronto, ON; Division of Gynecologic Oncology, Princess Margaret Cancer Centre, Toronto, ON
| | | | - Theodore J Brown
- The Lunenfeld-Tanenbaum Research Institute of Mount Sinai Hospital, Toronto, ON; Institute of Medical Sciences, University of Toronto, Toronto, ON; Department of Obstetrics and Gynecology, University of Toronto, Toronto, ON; Department of Physiology, University of Toronto, Toronto, ON.
| |
Collapse
|
47
|
Zhu J, Xin Y, Liu X, Wang Y, Liu Y. Nimotuzumab enhances the sensitivity of non-small cell lung cancer cells to tumor necrosis factor-α by inhibiting the nuclear factor-кB signaling pathway. Exp Ther Med 2018; 15:3345-3351. [PMID: 29545853 PMCID: PMC5841043 DOI: 10.3892/etm.2018.5856] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Accepted: 12/06/2017] [Indexed: 01/06/2023] Open
Abstract
Non-small cell lung cancer (NSCLC) accounts for ~85% of lung cancer cases worldwide. Current guidelines recommend the use of epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors for patients with NSCLC. The EGF/EGFR signaling pathway has been demonstrated to activate nuclear factor (NF)-κB, which may inhibit tumor necrosis factor (TNF)-α induced cell apoptosis. The aim of the present study was to investigate whether inhibiting the EGF/EGFR signaling pathway sensitizes NSCLC cell lines to TNF-α-induced apoptosis. The resistance of NSCLC cell lines to TNF-α was evaluated by cell viability assay. The effect of nimotuzumab (Ni) on NSCLC cell sensitivity to TNF-α, as well as the role of NF-κB in mediating resistance to TNF-α-induced apoptosis, was explored by western blot analysis, cell viability assay, apoptosis assay and an NF-κB DNA binding assay. It was demonstrated that EGFR protein expression was markedly higher in the H292 and H1975 cell lines compared with H460 and H1299 cell lines. H292 and H1975 also exhibited significantly increased TNF-α resistance compared with H460 and H1299 cells. Low dose Ni treatment slightly reduced the viability of H292 and H1975 cells; however, combined treatment with low dose Ni and TNF-α significantly inhibited H292 and H1299 cell viability compared with H460 and H1299 cells by inducing cell apoptosis. NF-κB protein expression and activity were also inhibited by the combination treatment. TNF-α treatment alone induced apoptosis in NF-κB deficient H292 and H1975 cells, similar to the effect of combination treatment in wild type H292 and H1975 cells. The results of the present study suggest that Ni sensitizes NSCLC cell lines to TNF-α-induced cell death by inhibiting NF-κB protein expression and activation, indicating a novel mechanism by which Ni suppresses the development of NSCLC.
Collapse
Affiliation(s)
- Jing Zhu
- Department of Thoracic Oncology, Cancer Hospital of Jilin Province, Changchun, Jilin 130012, P.R. China
| | - Ying Xin
- Department of Thoracic Oncology, Cancer Hospital of Jilin Province, Changchun, Jilin 130012, P.R. China
| | - Xiaoliang Liu
- Department of Blood Cancer, The First Affiliated Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Ying Wang
- Department of Thoracic Oncology, Cancer Hospital of Jilin Province, Changchun, Jilin 130012, P.R. China
| | - Ying Liu
- Department of Thoracic Oncology, Cancer Hospital of Jilin Province, Changchun, Jilin 130012, P.R. China
| |
Collapse
|
48
|
Woo SH, Seo SK, Park Y, Kim EK, Seong MK, Kim HA, Song JY, Hwang SG, Lee JK, Noh WC, Park IC. Dichloroacetate potentiates tamoxifen-induced cell death in breast cancer cells via downregulation of the epidermal growth factor receptor. Oncotarget 2018; 7:59809-59819. [PMID: 27494858 PMCID: PMC5312350 DOI: 10.18632/oncotarget.10999] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Accepted: 07/22/2016] [Indexed: 01/01/2023] Open
Abstract
Metabolic reprogramming in cancer cells has recently been recognized as an essential hallmark of neoplasia. In this context, metabolic alterations represent an attractive therapeutic target, and encouraging results with drugs targeting various metabolic processes have been obtained in preclinical studies. Recently, several studies have suggested that dichloroacetate (DCA), a specific pyruvate dehydrogenase kinase inhibitor, may be a potential anticancer drug in a large number of diverse tumors. However, the precise mechanism is not fully understood, which is important for the use of DCA in cancer treatment. In the present study, we found that DCA sensitized MCF7 breast cancer cells to tamoxifen-induced cell death by decreasing epidermal growth factor receptor (EGFR) expression. The downregulation of EGFR was caused by degradation of the protein. Furthermore, p38 mitogen-activated protein kinase played an important role in DCA/tamoxifen-induced EGFR degradation. Finally, DCA also promoted comparable tamoxifen-induced cell death in tamoxifen-resistant MCF7 cells, which were established by long-term treatment with tamoxifen. In summary, our results suggest that DCA is an attractive potential drug that sensitizes cells to tamoxifen-induced cell death and overcome tamoxifen resistance via downregulation of EGFR expression in breast cancer cells.
Collapse
Affiliation(s)
- Sang Hyeok Woo
- Division of Radiation Cancer Research, Korea Institute of Radiological and Medical Sciences, Nowon-gu, Seoul, 01812, Republic of Korea
| | - Sung-Keum Seo
- Division of Radiation Cancer Research, Korea Institute of Radiological and Medical Sciences, Nowon-gu, Seoul, 01812, Republic of Korea
| | - Yoonhwa Park
- Division of Radiation Cancer Research, Korea Institute of Radiological and Medical Sciences, Nowon-gu, Seoul, 01812, Republic of Korea.,School of Life Science and Biotechnology, Korea University, Seongbuk-gu, Seoul, 02841, Republic of Korea
| | - Eun-Kyu Kim
- Department of Surgery, Breast Cancer Center, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Bundang-gu, Seongnam, 13620, Republic of Korea
| | - Min-Ki Seong
- Department of Surgery, Korea Cancer Center Hospital, Korea Institute of Radiological and Medical Sciences, Nowon-gu, Seoul, 01812, Republic of Korea
| | - Hyun-Ah Kim
- Department of Surgery, Korea Cancer Center Hospital, Korea Institute of Radiological and Medical Sciences, Nowon-gu, Seoul, 01812, Republic of Korea
| | - Jie-Young Song
- Division of Radiation Cancer Research, Korea Institute of Radiological and Medical Sciences, Nowon-gu, Seoul, 01812, Republic of Korea
| | - Sang-Gu Hwang
- Division of Radiation Cancer Research, Korea Institute of Radiological and Medical Sciences, Nowon-gu, Seoul, 01812, Republic of Korea
| | - Jin Kyung Lee
- KIRAMS Radiation Biobank, Korea Institute of Radiological and Medical Sciences, Nowon-gu, Seoul, 01812, Republic of Korea
| | - Woo Chul Noh
- Department of Surgery, Korea Cancer Center Hospital, Korea Institute of Radiological and Medical Sciences, Nowon-gu, Seoul, 01812, Republic of Korea
| | - In-Chul Park
- Division of Radiation Cancer Research, Korea Institute of Radiological and Medical Sciences, Nowon-gu, Seoul, 01812, Republic of Korea
| |
Collapse
|
49
|
Tomkova S, Misuth M, Lenkavska L, Miskovsky P, Huntosova V. In vitro identification of mitochondrial oxidative stress production by time-resolved fluorescence imaging of glioma cells. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2018; 1865:616-628. [PMID: 29410069 DOI: 10.1016/j.bbamcr.2018.01.012] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Revised: 01/29/2018] [Accepted: 01/30/2018] [Indexed: 02/07/2023]
Abstract
Oxidative phosphorylation and glycolysis are important features, by which cells could bypass oxidative stress. The level of oxidative stress, and the ability of cells to promote oxidative phosphorylation or glycolysis, significantly determined proliferation or cell demise. In the present work, we have employed selective mitochondrial probe MitoTracker™ Orange CMTM/Ros (MTO) to estimate the level of oxidative stress in cancer cells at different stressed conditions. MTO is partially sensitive to decrease of mitochondrial membrane potential and to reactive oxygen species (ROS) generated in mitochondria. We have demonstrated, that fluorescence lifetime of MTO is much more sensitive to oxidative stress than intensity-based approaches. This method was validated in different cancer cell lines. Our approach revealed, at relatively low ROS levels, that Gö 6976, a protein kinase C (PKC) α inhibitor, and rottlerin, an indirect PKCδ inhibitor, increased mitochondrial ROS level in glioma cell. Their involvement in oxidative phosphorylation and apoptosis was investigated with oxygen consumption rate estimation, western blot and flow-cytometric analysis. Our study brings new insight to identify feeble differences in ROS production in living cells.
Collapse
Affiliation(s)
- Silvia Tomkova
- Department of Biophysics, Faculty of Science, P. J. Safarik University in Kosice, Jesenna 5, 041 54, Kosice, Slovakia
| | - Matus Misuth
- Department of Biophysics, Faculty of Science, P. J. Safarik University in Kosice, Jesenna 5, 041 54, Kosice, Slovakia
| | - Lenka Lenkavska
- Department of Biophysics, Faculty of Science, P. J. Safarik University in Kosice, Jesenna 5, 041 54, Kosice, Slovakia
| | - Pavol Miskovsky
- Center for Interdisciplinary Biosciences, Technology and innovation park, P.J. Safarik University in Kosice, Jesenna 5, 041 54, Kosice, Slovakia; SAFTRA photonics Ltd., Jesenna 5, 041 54, Kosice, Slovakia
| | - Veronika Huntosova
- Center for Interdisciplinary Biosciences, Technology and innovation park, P.J. Safarik University in Kosice, Jesenna 5, 041 54, Kosice, Slovakia.
| |
Collapse
|
50
|
Wang D, Tang H, Xu X, Dai W, Wu J, Wang J. Control the intracellular NF-κB activity by a sensor consisting of miRNA and decoy. Int J Biochem Cell Biol 2018; 95:43-52. [PMID: 29246684 DOI: 10.1016/j.biocel.2017.12.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2017] [Revised: 11/14/2017] [Accepted: 12/08/2017] [Indexed: 12/14/2022]
Abstract
Many diseases are associated with the abnormal activation of NF-κB and its signaling pathway. NF-κB has become an important target for disease treatment and development of new drugs. Many various NF-κB inhibitors were therefore developed; however, they have difficulties to become clinical drugs due to their adverse side effects resulted from the affected normal physiological functions of this transcription factor. To overcome this limitation, this study construct a transgenic vector that can express an artificial miRNA targeting NF-κB RelA under the control of a NF-κB-specific promoter. The promoter consists of a NF-κB decoy and a minimal promoter. The vector was named as decoy minimal promoter-artificial microRNA (DMP-amiRNA). This study verified that this vector can sense and control the intracellular NF-κB activity upon transfection. Working of the vector forms a perfect feedback loop that realizes the NF-κB self-control. With the vector in cells, the higher NF-κB activity, the higher DMP transcriptional activity, and the more amiR533 expression. DMP-amiRNA can moderately inhibit the intracellular NF-κB activity but exert no significant effect on cell viability. This study therefore develops a new strategy for inhibiting over activity of NF-κB, which should has great potential in clinical application.
Collapse
Affiliation(s)
- Danyang Wang
- State Key Laboratory of Bioelectronics, Southeast University, Nanjing 210096, China
| | - Huanhuan Tang
- State Key Laboratory of Bioelectronics, Southeast University, Nanjing 210096, China
| | - Xinhui Xu
- State Key Laboratory of Bioelectronics, Southeast University, Nanjing 210096, China
| | - Wei Dai
- State Key Laboratory of Bioelectronics, Southeast University, Nanjing 210096, China
| | - Jian Wu
- State Key Laboratory of Bioelectronics, Southeast University, Nanjing 210096, China
| | - Jinke Wang
- State Key Laboratory of Bioelectronics, Southeast University, Nanjing 210096, China.
| |
Collapse
|