1
|
Maleszewska M, Roura AJ, Dabrowski MJ, Draminski M, Wojtas B. Decoding glioblastoma's diversity: Are neurons part of the game? Cancer Lett 2025; 620:217666. [PMID: 40147584 DOI: 10.1016/j.canlet.2025.217666] [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: 01/04/2025] [Revised: 03/18/2025] [Accepted: 03/21/2025] [Indexed: 03/29/2025]
Abstract
Glioblastoma multiforme (GBM, WHO Grade 4) is a highly aggressive primary brain tumor with limited treatment options and a poor prognosis. A key challenge in GBM therapy lies in its pronounced heterogeneity, both within individual tumors (intratumoral) and between patients (intertumoral). Historically, neurons have been underexplored in GBM research; however, recent studies reveal that GBM development is closely linked to neural and glial progenitors, often mimicking neurodevelopmental processes in a dysregulated manner. Beyond damaging neuronal tissue, GBM actively engages with neurons to promote pro-tumorigenic signaling, including neuronal hyperexcitability and seizures. Single-cell RNA sequencing (scRNA-seq) has revolutionized our understanding of the tumor microenvironment (TME), uncovering the critical roles of immune cells, endothelial cells, and astrocytes in tumor progression. However, technical limitations of scRNA-seq hinder its ability to capture the transcriptomes of neurons, necessitating the use of single-nucleus RNA sequencing (snRNA-seq) to study these interactions at single-cell resolution. This work collects the emerging insights of glioblastoma-neuron interactions, focusing on how GBM exploits neurodevelopmental pathways and reshapes neuronal networks. Moreover, we perform bioinformatic analysis of publicly available snRNA-seq datasets to propose putative cell-cell interactions driving glioma-neuronal dynamics. This study delineates key signaling pathways and underscores the need for further investigation to evaluate their potential as therapeutic targets.
Collapse
Affiliation(s)
- Marta Maleszewska
- Department of Animal Physiology, Institute of Experimental Zoology, Faculty of Biology, University of Warsaw, 1 Miecznikowa Str, 02-096, Warsaw, Poland.
| | - Adrià-Jaume Roura
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain; Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Barcelona, Spain
| | - Michal J Dabrowski
- Computational Biology Group, Institute of Computer Science of the Polish Academy of Sciences, Warsaw, Poland
| | - Michal Draminski
- Computational Biology Group, Institute of Computer Science of the Polish Academy of Sciences, Warsaw, Poland
| | - Bartosz Wojtas
- Laboratory of Sequencing, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| |
Collapse
|
2
|
Kelly H, Inada M, Itoh Y. The Diverse Pathways for Cell Surface MT1-MMP Localization in Migratory Cells. Cells 2025; 14:209. [PMID: 39937000 PMCID: PMC11816416 DOI: 10.3390/cells14030209] [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: 10/22/2024] [Revised: 01/26/2025] [Accepted: 01/30/2025] [Indexed: 02/13/2025] Open
Abstract
Controlled cell migration is an essential biological process in health, while uncontrolled cell migration contributes to disease progression. For cells to migrate through tissue, they must first degrade the extracellular matrix (ECM), which acts as a physical barrier to cell migration. A type I transmembrane-type matrix metalloproteinase, MT1-MMP, is the key enzyme involved in this process. It has been extensively shown that MT1-MMP promotes the migration of different cell types in tissue, including fibroblasts, epithelial cells, endothelial cells, macrophages, mesenchymal stem cells, and cancer cells. MT1-MMP is tightly regulated at different levels, and its localization to leading-edge membrane structures is an essential process for MT1-MMP to promote cellular invasion. Different cells display different motility-associated membrane structures, which contribute to their invasive ability, and there are diverse mechanisms of MT1-MMP localization to these structures. In this article, we will discuss the current understanding of MT1-MMP regulation, in particular, localization mechanisms to these different motility-associated membrane structures.
Collapse
Affiliation(s)
- Hannah Kelly
- The Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford OX3 7FY, UK;
| | - Masaki Inada
- Department of Biotechnology and Life Science, Tokyo University of Agriculture and Technology, 2-24-16 Nakacho, Koganei, Tokyo 184-8588, Japan;
- Institute of Global Innovation Research, Tokyo University of Agriculture and Technology, 2-24-16 Nakacho, Koganei, Tokyo 184-8588, Japan
| | - Yoshifumi Itoh
- The Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford OX3 7FY, UK;
- Institute of Global Innovation Research, Tokyo University of Agriculture and Technology, 2-24-16 Nakacho, Koganei, Tokyo 184-8588, Japan
| |
Collapse
|
3
|
Zhao B, Ye DM, Li S, Zhang Y, Zheng Y, Kang J, Wang L, Zhao N, Ahmad B, Sun J, Yu T, Wu H. FMNL3 Promotes Migration and Invasion of Breast Cancer Cells via Inhibiting Rad23B-Induced Ubiquitination of Twist1. J Cell Physiol 2025; 240:e31481. [PMID: 39582466 DOI: 10.1002/jcp.31481] [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: 06/10/2024] [Revised: 10/18/2024] [Accepted: 10/24/2024] [Indexed: 11/26/2024]
Abstract
Breast cancer is a heterogeneous malignant tumor, and its high metastasis rate depends on the abnormal activation of cell dynamics. Formin-like protein 3 (FMNL3) plays an important role in the formation of various cytoskeletons that participate in cell movement. The objective of this study was to explore the function of FMNL3 in breast cancer progression and endeavor to reveal the molecular mechanism of this phenomenon. We found that FMNL3 was abnormally highly expressed in aggressive breast cancer cells and tissues, and it significantly inhibited E-cadherin expression. FMNL3 could specifically interact with Twist1 rather than other epithelial-mesenchymal transition transcription factors (EMT-TFs). We also found that FMNL3 enhanced the repressive effect of Twist1 on CDH1 transcription in breast cancer cells. Further mechanism studies showed that FMNL3 suppressed the ubiquitin degradation of Twist1 by inhibiting the interaction between Twist1 and Rad23B, the ubiquitin transfer protein of Twist1. In vitro functional experiments, it was confirmed that FMNL3 promoted the migration and invasion of breast cancer cells by regulating Twist1. Furthermore, Twist1 could directly bind to the fmnl3 promoter to facilitate FMNL3 transcription. To conclude, this study indicated that FMNL3 acted as a pro-metastasis factor in breast cancer by promoting Twist1 stability to suppress CDH1 transcription.
Collapse
Affiliation(s)
- Binggong Zhao
- School of Bioengineering and Key Laboratory of Protein Modification and Disease, Dalian University of Technology, Dalian, Liaoning, China
| | - Dong-Man Ye
- Department of Medical Imaging, Cancer Hospital of Dalian University of Technology, Shenyang, Liaoning, China
| | - Shujing Li
- School of Bioengineering and Key Laboratory of Protein Modification and Disease, Dalian University of Technology, Dalian, Liaoning, China
| | - Yong Zhang
- Department of Medical Imaging, Cancer Hospital of Dalian University of Technology, Shenyang, Liaoning, China
| | - Yang Zheng
- Department of Medical Imaging, Cancer Hospital of Dalian University of Technology, Shenyang, Liaoning, China
| | - Jie Kang
- School of Bioengineering and Key Laboratory of Protein Modification and Disease, Dalian University of Technology, Dalian, Liaoning, China
| | - Luhong Wang
- School of Bioengineering and Key Laboratory of Protein Modification and Disease, Dalian University of Technology, Dalian, Liaoning, China
| | - Nannan Zhao
- Department of Medical Imaging, Cancer Hospital of Dalian University of Technology, Shenyang, Liaoning, China
| | - Bashir Ahmad
- School of Bioengineering and Key Laboratory of Protein Modification and Disease, Dalian University of Technology, Dalian, Liaoning, China
| | - Jing Sun
- Department of Medical Imaging, Cancer Hospital of Dalian University of Technology, Shenyang, Liaoning, China
| | - Tao Yu
- Department of Medical Imaging, Cancer Hospital of Dalian University of Technology, Shenyang, Liaoning, China
| | - Huijian Wu
- School of Bioengineering and Key Laboratory of Protein Modification and Disease, Dalian University of Technology, Dalian, Liaoning, China
| |
Collapse
|
4
|
Nturubika BDD, Logan J, Johnson IRD, Moore C, Li KL, Tang J, Lam G, Parkinson-Lawrence E, Williams DB, Chakiris J, Hindes M, Brooks RD, Miles MA, Selemidis S, Gregory P, Weigert R, Butler L, Ward MP, Waugh DJJ, O’Leary JJ, Brooks DA. Components of the Endosome-Lysosome Vesicular Machinery as Drivers of the Metastatic Cascade in Prostate Cancer. Cancers (Basel) 2024; 17:43. [PMID: 39796673 PMCID: PMC11718918 DOI: 10.3390/cancers17010043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 12/16/2024] [Accepted: 12/22/2024] [Indexed: 01/13/2025] Open
Abstract
Prostate cancer remains a significant global health concern, with over 1.4 million new cases diagnosed and more than 330,000 deaths each year. The primary clinical challenge that contributes to poor patient outcomes involves the failure to accurately predict and treat at the onset of metastasis, which remains an incurable stage of the disease. This review discusses the emerging paradigm that prostate cancer metastasis is driven by a dysregulation of critical molecular machinery that regulates endosome-lysosome homeostasis. Endosome and lysosome compartments have crucial roles in maintaining normal cellular function but are also involved in many hallmarks of cancer pathogenesis, including inflammation, immune response, nutrient sensing, metabolism, proliferation, signalling, and migration. Here we discuss new insight into how alterations in the complex network of trafficking machinery, responsible for the microtubule-based transport of endosomes and lysosomes, may be involved in prostate cancer progression. A better understanding of endosome-lysosome dynamics may facilitate the discovery of novel strategies to detect and manage prostate cancer metastasis and improve patient outcomes.
Collapse
Affiliation(s)
- Bukuru Dieu-Donne Nturubika
- Clinical and Health Sciences, University of South Australia, Adelaide, SA 5000, Australia; (J.L.); (C.M.); (K.L.L.); (J.T.); (E.P.-L.); (D.B.W.); (J.C.); (M.H.); (R.D.B.)
| | - Jessica Logan
- Clinical and Health Sciences, University of South Australia, Adelaide, SA 5000, Australia; (J.L.); (C.M.); (K.L.L.); (J.T.); (E.P.-L.); (D.B.W.); (J.C.); (M.H.); (R.D.B.)
| | - Ian R. D. Johnson
- Clinical and Health Sciences, University of South Australia, Adelaide, SA 5000, Australia; (J.L.); (C.M.); (K.L.L.); (J.T.); (E.P.-L.); (D.B.W.); (J.C.); (M.H.); (R.D.B.)
| | - Courtney Moore
- Clinical and Health Sciences, University of South Australia, Adelaide, SA 5000, Australia; (J.L.); (C.M.); (K.L.L.); (J.T.); (E.P.-L.); (D.B.W.); (J.C.); (M.H.); (R.D.B.)
| | - Ka Lok Li
- Clinical and Health Sciences, University of South Australia, Adelaide, SA 5000, Australia; (J.L.); (C.M.); (K.L.L.); (J.T.); (E.P.-L.); (D.B.W.); (J.C.); (M.H.); (R.D.B.)
| | - Jingying Tang
- Clinical and Health Sciences, University of South Australia, Adelaide, SA 5000, Australia; (J.L.); (C.M.); (K.L.L.); (J.T.); (E.P.-L.); (D.B.W.); (J.C.); (M.H.); (R.D.B.)
| | - Giang Lam
- Centre for Cancer Biology, University of South Australia, Adelaide, SA 5000, Australia; (G.L.); (P.G.); (D.J.J.W.)
| | - Emma Parkinson-Lawrence
- Clinical and Health Sciences, University of South Australia, Adelaide, SA 5000, Australia; (J.L.); (C.M.); (K.L.L.); (J.T.); (E.P.-L.); (D.B.W.); (J.C.); (M.H.); (R.D.B.)
| | - Desmond B. Williams
- Clinical and Health Sciences, University of South Australia, Adelaide, SA 5000, Australia; (J.L.); (C.M.); (K.L.L.); (J.T.); (E.P.-L.); (D.B.W.); (J.C.); (M.H.); (R.D.B.)
| | - James Chakiris
- Clinical and Health Sciences, University of South Australia, Adelaide, SA 5000, Australia; (J.L.); (C.M.); (K.L.L.); (J.T.); (E.P.-L.); (D.B.W.); (J.C.); (M.H.); (R.D.B.)
| | - Madison Hindes
- Clinical and Health Sciences, University of South Australia, Adelaide, SA 5000, Australia; (J.L.); (C.M.); (K.L.L.); (J.T.); (E.P.-L.); (D.B.W.); (J.C.); (M.H.); (R.D.B.)
| | - Robert D. Brooks
- Clinical and Health Sciences, University of South Australia, Adelaide, SA 5000, Australia; (J.L.); (C.M.); (K.L.L.); (J.T.); (E.P.-L.); (D.B.W.); (J.C.); (M.H.); (R.D.B.)
| | - Mark A. Miles
- Centre for Respiratory Science and Health, School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC 3083, Australia; (M.A.M.); (S.S.)
| | - Stavros Selemidis
- Centre for Respiratory Science and Health, School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC 3083, Australia; (M.A.M.); (S.S.)
| | - Philip Gregory
- Centre for Cancer Biology, University of South Australia, Adelaide, SA 5000, Australia; (G.L.); (P.G.); (D.J.J.W.)
| | - Roberto Weigert
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA;
| | - Lisa Butler
- South Australian ImmunoGENomics Cancer Institute, Freemasons Centre for Male Health and Wellbeing, University of Adelaide, Adelaide, SA 5000, Australia;
- Solid Tumour Program, Precision Cancer Medicine Theme, South Australian Health and Medical Research Institute, Adelaide, SA 5000, Australia
| | - Mark P. Ward
- Department of Pathology, The Coombe Women and Infants University Hospital, Trinity College Dublin, D08 XW7X Dublin, Ireland;
| | - David J. J. Waugh
- Centre for Cancer Biology, University of South Australia, Adelaide, SA 5000, Australia; (G.L.); (P.G.); (D.J.J.W.)
| | - John J. O’Leary
- Department of Histopathology, Trinity College Dublin, D08 XW7X Dublin, Ireland;
| | - Douglas A. Brooks
- Clinical and Health Sciences, University of South Australia, Adelaide, SA 5000, Australia; (J.L.); (C.M.); (K.L.L.); (J.T.); (E.P.-L.); (D.B.W.); (J.C.); (M.H.); (R.D.B.)
- Department of Histopathology, Trinity College Dublin, D08 XW7X Dublin, Ireland;
| |
Collapse
|
5
|
Golmohammadi M, Zamanian MY, Al‐Ani AM, Jabbar TL, Kareem AK, Aghaei ZH, Tahernia H, Hjazi A, Jissir SA, Hakimizadeh E. Targeting STAT3 signaling pathway by curcumin and its analogues for breast cancer: A narrative review. Animal Model Exp Med 2024; 7:853-867. [PMID: 39219410 PMCID: PMC11680487 DOI: 10.1002/ame2.12491] [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: 06/15/2024] [Accepted: 08/10/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND Breast cancer (BC) continues to be a significant global health issue, with a rising number of cases requiring ongoing research and innovation in treatment strategies. Curcumin (CUR), a natural compound derived from Curcuma longa, and similar compounds have shown potential in targeting the STAT3 signaling pathway, which plays a crucial role in BC progression. AIMS The aim of this study was to investigate the effects of curcumin and its analogues on BC based on cellular and molecular mechanisms. MATERIALS & METHODS The literature search conducted for this study involved utilizing the Scopus, ScienceDirect, PubMed, and Google Scholar databases in order to identify pertinent articles. RESULTS This narrative review explores the potential of CUR and similar compounds in inhibiting STAT3 activation, thereby suppressing the proliferation of cancer cells, inducing apoptosis, and inhibiting metastasis. The review demonstrates that CUR directly inhibits the phosphorylation of STAT3, preventing its movement into the nucleus and its ability to bind to DNA, thereby hindering the survival and proliferation of cancer cells. CUR also enhances the effectiveness of other therapeutic agents and modulates the tumor microenvironment by affecting tumor-associated macrophages (TAMs). CUR analogues, such as hydrazinocurcumin (HC), FLLL11, FLLL12, and GO-Y030, show improved bioavailability and potency in inhibiting STAT3, resulting in reduced cell proliferation and increased apoptosis. CONCLUSION CUR and its analogues hold promise as effective adjuvant treatments for BC by targeting the STAT3 signaling pathway. These compounds provide new insights into the mechanisms of action of CUR and its potential to enhance the effectiveness of BC therapies.
Collapse
Affiliation(s)
| | - Mohammad Yassin Zamanian
- Department of Physiology, School of MedicineHamadan University of Medical SciencesHamadanIran
- Department of Pharmacology and Toxicology, School of PharmacyHamadan University of Medical SciencesHamadanIran
| | - Ahmed Muzahem Al‐Ani
- Department of Medical Laboratories TechnologyAL‐Nisour University CollegeBaghdadIraq
| | | | - Ali Kamil Kareem
- Biomedical Engineering DepartmentAl‐Mustaqbal University CollegeHillahIraq
| | - Zeinab Hashem Aghaei
- Preventative Gynecology Research CenterShahid Beheshti University of Medical SciencesTehranIran
| | - Hossein Tahernia
- Molecular Medicine Research Center, Research Institute of Basic Medical SciencesRafsanjan University of Medical SciencesRafsanjanIran
| | - Ahmed Hjazi
- Department of Medical Laboratory Sciences, College of Applied Medical SciencesPrince Sattam bin Abdulaziz UniversityAl‐KharjSaudi Arabia
| | | | - Elham Hakimizadeh
- Physiology‐Pharmacology Research Center, Research Institute of Basic Medical SciencesRafsanjan University of Medical SciencesRafsanjanIran
| |
Collapse
|
6
|
Dai B, Xu L, Rong S, Song M, Lan Z, Chen W, Zhang L, Liu Y, Wang L, Li J, Chen J, Wu Z. YTHDF2 promotes anaplastic thyroid cancer progression by activating the DDIT4/AKT/mTOR signaling pathway. Biol Direct 2024; 19:122. [PMID: 39593172 PMCID: PMC11600618 DOI: 10.1186/s13062-024-00566-y] [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: 10/10/2024] [Accepted: 11/07/2024] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND RNA methylation, an important reversible post-transcriptional modification in eukaryotes, has emerged as a prevalent epigenetic alteration. However, the role of the m6A reader YTH domain family 2 (YTHDF2) has not been reported in anaplastic thyroid cancer (ATC) and its biological mechanism is unclear. METHODS The relationship between YTHDF2 expression and ATC was determined using data sets and tissue samples. A range of analytical techniques were employed to investigate the regulatory mechanism of YTHDF2 in ATC, including bioinformatics analysis, m6A dot-blot analysis, methylated RNA immunoprecipitation sequencing (MeRIP-seq), RNA immunoprecipitation (RIP) assays, RNA sequencing, RNA stability assays and dual luciferase reporter gene assays. In vitro and in vivo assays were also conducted to determine the contribution of YTHDF2 to ATC development. RESULTS YTHDF2 expression was significantly increased in ATC. The comprehensive in vitro and in vivo experiments demonstrated that YTHDF2 knockdown significantly attenuated ATC proliferation, invasion, migration, and apoptosis promotion, whereas YTHDF2 overexpression yielded the opposite trend. Mechanistically, RNA-seq, MeRIP-seq and RIP-seq analysis, and molecular biology experiments demonstrated that YTHDF2 accelerated the degradation of DNA damage-inducible transcript 4 or regulated in DNA damage and development 1 (DDIT4, or REDD1) mRNA in an m6A-dependent manner, which in turn activated the AKT/mTOR signaling pathway and induced activation of epithelial-mesenchymal transition (EMT), thereby promoting ATC tumor progression. CONCLUSIONS This study is the first to demonstrate that elevated YTHDF2 expression levels suppress DDIT4 expression in an m6A-dependent manner and activate the AKT/mTOR signaling pathway, thereby promoting ATC progression. YTHDF2 plays a pivotal role in ATC progression, and it may serve as a promising therapeutic target in the future.
Collapse
Affiliation(s)
- Bao Dai
- Department of Thyroid and Hernia Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, Guangdong, 510080, China
| | - Lei Xu
- Department of Thyroid and Hernia Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, Guangdong, 510080, China
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Shikuo Rong
- Division of Thyroid surgery, Department of General Surgery, Shenzhen People's Hospital (The First Affiliated Hospital, Southern University of Science and Technology; The Second Clinical Medical College, Jinan University ), Shenzhen, Guangdong, China
| | - Muye Song
- Department of Anesthesiology, Guangdong Provincial People's Hospital Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, Guangdong, 510080, China
| | - Ziteng Lan
- Department of Thyroid and Hernia Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, Guangdong, 510080, China
| | - Weijian Chen
- Department of Thyroid and Hernia Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, Guangdong, 510080, China
| | - Lingyun Zhang
- Department of Thyroid and Hernia Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, Guangdong, 510080, China
| | - Yongchen Liu
- Department of Thyroid and Hernia Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, Guangdong, 510080, China
| | - Linhe Wang
- Department of Thyroid and Hernia Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, Guangdong, 510080, China
| | - Jinghua Li
- Department of Laboratory, Guangdong Provincial People's Hospital Guangdong Academy of Medical Sciences, Southern Medical University, 106 Zhongshan 2nd Road, Guangzhou, Guangdong, 510080, China.
| | - Jian Chen
- Department of Thyroid and Hernia Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, Guangdong, 510080, China.
| | - Zeyu Wu
- Department of Thyroid and Hernia Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, Guangdong, 510080, China.
| |
Collapse
|
7
|
Coli A, Gao S, Kaestner L. Sodium-Selective Channelrhodopsins. Cells 2024; 13:1852. [PMID: 39594600 PMCID: PMC11592924 DOI: 10.3390/cells13221852] [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: 09/23/2024] [Revised: 11/04/2024] [Accepted: 11/06/2024] [Indexed: 11/28/2024] Open
Abstract
Channelrhodopsins (ChRs) are light-gated ion channels originally discovered in algae and are commonly used in neuroscience for controlling the electrical activity of neurons with high precision. Initially-discovered ChRs were non-selective cation channels, allowing the flow of multiple ions, such as Na+, K+, H+, and Ca2+, leading to membrane depolarization and triggering action potentials in neurons. As the field of optogenetics has evolved, ChRs with more specific ion selectivity were discovered or engineered, offering more precise optogenetic manipulation. This review highlights the natural occurrence and engineered variants of sodium-selective channelrhodopsins (NaChRs), emphasizing their importance in optogenetic applications. These tools offer enhanced specificity in Na+ ion conduction, reducing unwanted effects from other ions, and generating strong depolarizing currents. Some of the NaChRs showed nearly no desensitization upon light illumination. These characteristics make them particularly useful for experiments requiring robust depolarization or direct Na+ ion manipulation. The review further discusses the molecular structure of these channels, recent advances in their development, and potential applications, including a proposed drug delivery system using NaChR-expressing red blood cells that could be triggered to release therapeutic agents upon light activation. This review concludes with a forward-looking perspective on expanding the use of NaChRs in both basic research and clinical settings.
Collapse
Affiliation(s)
- Ariel Coli
- Dynamics of Fluids, Experimental Physics, Saarland University, 66123 Saarbrücken, Germany;
| | - Shiqiang Gao
- Department of Neurophysiology, Physiological Institute, University of Würzburg, 97070 Würzburg, Germany;
| | - Lars Kaestner
- Dynamics of Fluids, Experimental Physics, Saarland University, 66123 Saarbrücken, Germany;
- Theoretical Medicine and Biosciences, Medical Faculty, Saarland University, 66421 Homburg, Germany
| |
Collapse
|
8
|
Sun L, Liu L, Jiang J, Liu K, Zhu J, Wu L, Lu X, Huang Z, Yuan Y, Crowley SD, Mao H, Xing C, Ren J. Transcription factor Twist1 drives fibroblast activation to promote kidney fibrosis via signaling proteins Prrx1/TNC. Kidney Int 2024; 106:840-855. [PMID: 39181396 DOI: 10.1016/j.kint.2024.07.028] [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: 11/30/2023] [Revised: 07/12/2024] [Accepted: 07/26/2024] [Indexed: 08/27/2024]
Abstract
The transcription factor Twist1 plays a vital role in normal development in many tissue systems and continues to be important throughout life. However, inappropriate Twist1 activity has been associated with kidney injury and fibrosis, though the underlying mechanisms involved remain incomplete. Here, we explored the role of Twist1 in regulating fibroblast behaviors and the development kidney fibrosis. Initially Twist1 protein and activity was found to be markedly increased within interstitial myofibroblasts in fibrotic kidneys in both humans and rodents. Treatment of rat kidney interstitial fibroblasts with transforming growth factor-β1 (a profibrotic factor) also induced Twist1 expression in vitro. Gain- and loss-of-function experiments supported that Twist1 signaling was responsible for transforming growth factor-β1-induced fibroblast activation and fetal bovine serum-induced fibroblast proliferation. Mechanistically, Twist1 protein promoted kidney fibroblast activation by driving the expression of downstream signaling proteins, Prrx1 and Tnc. Twist1 directly enhanced binding to the promoter of Prrx1 but not TNC, whereas the promoter of TNC was directly bound by Prrx1. Finally, mice with fibroblast-specific deletion of Twist1 exhibited less Prrx1 and TNC protein abundance, interstitial extracellular matrix deposition and kidney inflammation in both the unilateral ureteral obstruction and ischemic-reperfusion injury-induced-kidney fibrotic models. Inhibition of Twist1 signaling with Harmine, a β-carboline alkaloid, improved extracellular matrix deposition in both injury models. Thus, our results suggest that Twist1 signaling promotes the activation and proliferation of kidney fibroblasts, contributing to the development of interstitial fibrosis, offering a potential therapeutic target for chronic kidney disease.
Collapse
Affiliation(s)
- Lianqin Sun
- Department of Nephrology, the First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Lishan Liu
- Department of Nephrology, the First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Juanjuan Jiang
- Department of Nephrology, the First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Kang Liu
- Department of Nephrology, the First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Jingfeng Zhu
- Department of Nephrology, the First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Lin Wu
- Department of Nephrology, the First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Xiaohan Lu
- Division of Nephrology, Department of Medicine, Duke University Medical Center, Durham, North Carolina, USA
| | - Zhimin Huang
- Department of Nephrology, the First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Yanggang Yuan
- Department of Nephrology, the First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Steven D Crowley
- Division of Nephrology, Department of Medicine, Duke University Medical Center, Durham, North Carolina, USA; Department of Medicine, Durham VA Medical Center, Durham, North Carolina, USA
| | - Huijuan Mao
- Department of Nephrology, the First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing, Jiangsu Province, China.
| | - Changying Xing
- Department of Nephrology, the First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing, Jiangsu Province, China.
| | - Jiafa Ren
- Department of Nephrology, the First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing, Jiangsu Province, China.
| |
Collapse
|
9
|
Haynes NM, Chadwick TB, Parker BS. The complexity of immune evasion mechanisms throughout the metastatic cascade. Nat Immunol 2024; 25:1793-1808. [PMID: 39285252 DOI: 10.1038/s41590-024-01960-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Accepted: 08/12/2024] [Indexed: 09/29/2024]
Abstract
Metastasis, the spread of cancer from a primary site to distant organs, is an important challenge in oncology. This Review explores the complexities of immune escape mechanisms used throughout the metastatic cascade to promote tumor cell dissemination and affect organotropism. Specifically, we focus on adaptive plasticity of disseminated epithelial tumor cells to understand how they undergo phenotypic transitions to survive microenvironmental conditions encountered during metastasis. The interaction of tumor cells and their microenvironment is analyzed, highlighting the local and systemic effects that innate and adaptive immune systems have in shaping an immunosuppressive milieu to foster aggressive metastatic tumors. Effectively managing metastatic disease demands a multipronged approach to target the parallel and sequential mechanisms that suppress anti-tumor immunity. This management necessitates a deep understanding of the complex interplay between tumor cells, their microenvironment and immune responses that we provide with this Review.
Collapse
Affiliation(s)
- Nicole M Haynes
- Cancer Evolution and Metastasis Program, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia
| | - Thomas B Chadwick
- Cancer Evolution and Metastasis Program, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia
| | - Belinda S Parker
- Cancer Evolution and Metastasis Program, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia.
| |
Collapse
|
10
|
Lebel M, Cliche DO, Charbonneau M, Brochu-Gaudreau K, Adam D, Brochiero E, Dubois CM, Cantin AM. Hypoxia Promotes Invadosome Formation by Lung Fibroblasts. Cells 2024; 13:1152. [PMID: 38995003 PMCID: PMC11240699 DOI: 10.3390/cells13131152] [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: 06/21/2024] [Accepted: 07/02/2024] [Indexed: 07/13/2024] Open
Abstract
Lung parenchymal hypoxia has emerged as a cardinal feature of idiopathic pulmonary fibrosis (IPF). Hypoxia promotes cancer cell invasion and metastasis through signaling that is dependent upon the lysophosphatidic acid (LPA) receptor, LPA1 (LPAR1). Abundant data indicate that LPA1-dependent signaling also enhances lung fibrogenesis in IPF. We recently reported that fibroblasts isolated from the lungs of individuals with IPF have an increased capacity to form subcellular matrix-degradative structures known as invadosomes, an event that correlates with the degree of lung fibrosis. We therefore hypothesized that hypoxia promotes invadosome formation in lung fibroblasts through LPA1-dependent signaling. Here, it is demonstrated that invadosome formation by fibroblasts from the lungs of individuals with advanced IPF is inhibited by both the tyrosine receptor kinase inhibitor nintedanib and inhibition of LPA1. In addition, exposure of normal human lung fibroblasts to either hypoxia or LPA increased their ability to form invadosomes. Mechanistically, the hypoxia-induced invadosome formation by lung fibroblasts was found to involve LPA1 and PDGFR-Akt signaling. We concluded that hypoxia increases the formation of invadosomes in lung fibroblasts through the LPA1 and PDGFR-Akt signaling axis, which represents a potential target for suppressing lung fibrosis.
Collapse
Affiliation(s)
- Mégane Lebel
- Respiratory Division, Department of Medicine, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada; (M.L.); (D.O.C.); (A.M.C.)
| | - Dominic O. Cliche
- Respiratory Division, Department of Medicine, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada; (M.L.); (D.O.C.); (A.M.C.)
| | - Martine Charbonneau
- Department of Immunology and Cell Biology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, 3001, 12 Avenue Nord, Sherbrooke, QC J1H 5N4, Canada; (M.C.); (K.B.-G.)
| | - Karine Brochu-Gaudreau
- Department of Immunology and Cell Biology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, 3001, 12 Avenue Nord, Sherbrooke, QC J1H 5N4, Canada; (M.C.); (K.B.-G.)
| | - Damien Adam
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC H2X 0A9, Canada; (D.A.); (E.B.)
| | - Emmanuelle Brochiero
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC H2X 0A9, Canada; (D.A.); (E.B.)
- Department of Medicine, Université de Montréal, Montréal, QC H3T 1J4, Canada
| | - Claire M. Dubois
- Department of Immunology and Cell Biology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, 3001, 12 Avenue Nord, Sherbrooke, QC J1H 5N4, Canada; (M.C.); (K.B.-G.)
| | - André M. Cantin
- Respiratory Division, Department of Medicine, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada; (M.L.); (D.O.C.); (A.M.C.)
| |
Collapse
|
11
|
Wu Y, Chen R, Ni S, Hu K. Biomimetic "nano-spears" for CAFs-targeting: splintered three "shields" with enhanced cisplatin anti-TNBC efficiency. J Control Release 2024; 370:556-569. [PMID: 38697316 DOI: 10.1016/j.jconrel.2024.04.053] [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: 01/26/2024] [Revised: 04/12/2024] [Accepted: 04/29/2024] [Indexed: 05/04/2024]
Abstract
The treatment dilemma of triple-negative breast cancer (TNBC) revolves around drug resistance and metastasis. Cancer-associated fibroblasts (CAFs) contribute to cisplatin (Cis) resistance and further metastasis in TNBC, making TNBC a difficult-to-treat disease. The dense stromal barrier which restricts drug delivery, invasive phenotype of tumor cells, and immunosuppressive tumor microenvironment (TME) induced by CAFs serve as three "shields" for TNBC against Cis therapy. Here, we designed a silybin-loaded biomimetic nanoparticle coated with anisamide-modified red blood cell membrane (ARm@SNP) as a "nanospear" for CAFs-targeting, which could shatter the "shields" and significantly exhibit inhibitory effect on 4T1 cells in combination with Cis both in vitro and in vivo. The ARm@SNP/Cis elicited 4T1 tumor growth arrest and destroyed three "shields" as follows: disintegrating the stromal barrier by inhibiting blood vessels growth and the expression of fibronectin; decreasing 4T1 cell invasion and metastasis by affecting the TGF-β/Twist/EMT pathway which impeded EMT activation; reversing the immunosuppressive microenvironment by increasing the activity and infiltration of immunocompetent cells. Based on CAFs-targeting, ARm@SNP reversed the resistance of Cis, remodeled the TME and inhibited invasion and metastasis while significantly improving the therapeutic effect of Cis on 4T1 tumor-bearing mice, providing a promising approach for treating intractable TNBC.
Collapse
Affiliation(s)
- Yufan Wu
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Rujing Chen
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Department of Pharmacy, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, China
| | - Shuting Ni
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Kaili Hu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| |
Collapse
|
12
|
Yu X, Xu J. TWIST1 Drives Cytotoxic CD8+ T-Cell Exhaustion through Transcriptional Activation of CD274 (PD-L1) Expression in Breast Cancer Cells. Cancers (Basel) 2024; 16:1973. [PMID: 38893094 PMCID: PMC11171171 DOI: 10.3390/cancers16111973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 05/15/2024] [Accepted: 05/18/2024] [Indexed: 06/21/2024] Open
Abstract
In breast cancer, epithelial-mesenchymal transition (EMT) is positively associated with programmed death ligand 1 (PD-L1) expression and immune escape, and TWIST1 silences ERα expression and induces EMT and cancer metastasis. However, how TWIST1 regulates PD-L1 and immune evasion is unknown. This study analyzed TWIST1 and PD-L1 expression in breast cancers, investigated the mechanism for TWIST1 to regulate PD-L1 transcription, and assessed the effects of TWIST1 and PD-L1 in cancer cells on cytotoxic CD8+ T cells. Interestingly, TWIST1 expression is correlated with high-level PD-L1 expression in ERα-negative breast cancer cells. The overexpression and knockdown of TWIST1 robustly upregulate and downregulate PD-L1 expression, respectively. TWIST1 binds to the PD-L1 promoter and recruits the TIP60 acetyltransferase complex in a BRD8-dependent manner to transcriptionally activate PD-L1 expression, which significantly accelerates the exhaustion and death of the cytotoxic CD8+ T cells. Accordingly, knockdown of TWIST1 or BRD8 or inhibition of PD-L1 significantly enhances the tumor antigen-specific CD8+ T cells to suppress the growth of breast cancer cells. These results demonstrate that TWIST1 directly induces PD-L1 expression in ERα-negative breast cancer cells to promote immune evasion. Targeting TWIST1, BRD8, and/or PD-L1 in ERα-negative breast cancer cells with TWIST1 expression may sensitize CD8+ T-cell-mediated immunotherapy.
Collapse
Affiliation(s)
- Xiaobin Yu
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA;
| | - Jianming Xu
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA;
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
13
|
Xue JD, Xiang WF, Cai MQ, Lv XY. Biological functions and therapeutic potential of SRY related high mobility group box 5 in human cancer. Front Oncol 2024; 14:1332148. [PMID: 38835366 PMCID: PMC11148273 DOI: 10.3389/fonc.2024.1332148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Accepted: 04/26/2024] [Indexed: 06/06/2024] Open
Abstract
Cancer is a heavy human burden worldwide, with high morbidity and mortality. Identification of novel cancer diagnostic and prognostic biomarkers is important for developing cancer treatment strategies and reducing mortality. Transcription factors, including SRY associated high mobility group box (SOX) proteins, are thought to be involved in the regulation of specific biological processes. There is growing evidence that SOX transcription factors play an important role in cancer progression, including tumorigenesis, changes in the tumor microenvironment, and metastasis. SOX5 is a member of SOX Group D of Sox family. SOX5 is expressed in various tissues of human body and participates in various physiological and pathological processes and various cellular processes. However, the abnormal expression of SOX5 is associated with cancer of various systems, and the abnormal expression of SOX5 acts as a tumor promoter to promote cancer cell viability, proliferation, invasion, migration and EMT through multiple mechanisms. In addition, the expression pattern of SOX5 is closely related to cancer type, stage and adverse clinical outcome. Therefore, SOX5 is considered as a potential biomarker for cancer diagnosis and prognosis. In this review, the expression of SOX5 in various human cancers, the mechanism of action and potential clinical significance of SOX5 in tumor, and the therapeutic significance of Sox5 targeting in cancer were reviewed. In order to provide a new theoretical basis for cancer clinical molecular diagnosis, molecular targeted therapy and scientific research.
Collapse
Affiliation(s)
- Juan-di Xue
- The School of Basic Medicine Sciences of Lanzhou University, Lanzhou, China
| | - Wan-Fang Xiang
- School/Hospital of Stomatology of Lanzhou University, Lanzhou, China
| | - Ming-Qin Cai
- School/Hospital of Stomatology of Lanzhou University, Lanzhou, China
| | - Xiao-Yun Lv
- The School of Basic Medicine Sciences of Lanzhou University, Lanzhou, China
| |
Collapse
|
14
|
Rea M, Kimmerer G, Mittendorf S, Xiong X, Green M, Chandler D, Saintilnord W, Blackburn J, Gao T, Fondufe-Mittendorf YN. A dynamic model of inorganic arsenic-induced carcinogenesis reveals an epigenetic mechanism for epithelial-mesenchymal plasticity. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 347:123586. [PMID: 38467368 PMCID: PMC11005477 DOI: 10.1016/j.envpol.2024.123586] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 02/12/2024] [Accepted: 02/14/2024] [Indexed: 03/13/2024]
Abstract
Inorganic arsenic (iAs) causes cancer by initiating dynamic transitions between epithelial and mesenchymal cell phenotypes. These transitions transform normal cells into cancerous cells, and cancerous cells into metastatic cells. Most in vitro models assume that transitions between states are binary and complete, and do not consider the possibility that intermediate, stable cellular states might exist. In this paper, we describe a new, two-hit in vitro model of iAs-induced carcinogenesis that extends to 28 weeks of iAs exposure. Through week 17, the model faithfully recapitulates known and expected phenotypic, genetic, and epigenetic characteristics of iAs-induced carcinogenesis. By 28 weeks, however, exposed cells exhibit stable, intermediate phenotypes and epigenetic properties, and key transcription factor promoters (SNAI1, ZEB1) enter an epigenetically poised or bivalent state. These data suggest that key epigenetic transitions and cellular states exist during iAs-induced epithelial-to-mesenchymal transition (EMT), and that it is important for our in vitro models to encapsulate all aspects of EMT and the mesenchymal-to-epithelial transition (MET). In so doing, and by understanding the epigenetic systems controlling these transitions, we might find new, unexpected opportunities for developing targeted, cell state-specific therapeutics.
Collapse
Affiliation(s)
- Matthew Rea
- Department of Epigenetics, Van Andel Institute, Grand Rapids, MI, 49502, USA
| | - Greg Kimmerer
- Department of Biology, University of Kentucky, Lexington, KY, 40506, USA
| | - Shania Mittendorf
- Department of Biology, University of Kentucky, Lexington, KY, 40506, USA
| | - Xiaopeng Xiong
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY, 40536, USA; Markey Cancer Center, University of Kentucky, Lexington, KY, 40536, USA
| | - Meghan Green
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY, 40536, USA; Markey Cancer Center, University of Kentucky, Lexington, KY, 40536, USA
| | - Darrell Chandler
- Department of Epigenetics, Van Andel Institute, Grand Rapids, MI, 49502, USA
| | - Wesley Saintilnord
- Department of Epigenetics, Van Andel Institute, Grand Rapids, MI, 49502, USA; Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY, 40536, USA
| | - Jessica Blackburn
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY, 40536, USA; Markey Cancer Center, University of Kentucky, Lexington, KY, 40536, USA
| | - Tianyan Gao
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY, 40536, USA; Markey Cancer Center, University of Kentucky, Lexington, KY, 40536, USA
| | | |
Collapse
|
15
|
Gil-Henn H, Girault JA, Lev S. PYK2, a hub of signaling networks in breast cancer progression. Trends Cell Biol 2024; 34:312-326. [PMID: 37586982 DOI: 10.1016/j.tcb.2023.07.006] [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: 05/01/2023] [Revised: 07/17/2023] [Accepted: 07/18/2023] [Indexed: 08/18/2023]
Abstract
Breast cancer (BC) involves complex signaling networks characterized by extensive cross-communication and feedback loops between and within multiple signaling cascades. Many of these signaling pathways are driven by genetic alterations of oncogene and/or tumor-suppressor genes and are influenced by various environmental cues. We describe unique roles of the non-receptor tyrosine kinase (NRTK) PYK2 in signaling integration and feedback looping in BC. PYK2 functions as a signaling hub in various cascades, and its involvement in positive and negative feedback loops enhances signaling robustness, modulates signaling dynamics, and contributes to BC growth, epithelial-to-mesenchymal transition (EMT), stemness, migration, invasion, and metastasis. We also discuss the potential of PYK2 as a therapeutic target in various BC subtypes.
Collapse
Affiliation(s)
- Hava Gil-Henn
- The Azrieli Faculty of Medicine, Bar-Ilan University, Safed 1311502, Israel
| | - Jean-Antoine Girault
- Institut du Fer à Moulin, Institut National de la Santé et de la Recherche Médicale (INSERM) Unité Mixte de Recherche en Santé (UMRS) 1270, Sorbonne Université, 75005 Paris, France
| | - Sima Lev
- Molecular Cell Biology Department, Weizmann Institute of Science, Rehovot 76100, Israel.
| |
Collapse
|
16
|
Mahdi-Esferizi R, Shiasi Z, Heidari R, Najafi A, Mahmoudi I, Elahian F, Tahmasebian S. Single-cell transcriptional signature-based drug repurposing and in vitro evaluation in colorectal cancer. BMC Cancer 2024; 24:371. [PMID: 38528462 DOI: 10.1186/s12885-024-12142-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Accepted: 03/18/2024] [Indexed: 03/27/2024] Open
Abstract
BACKGROUND The need for intelligent and effective treatment of diseases and the increase in drug design costs have raised drug repurposing as one of the effective strategies in biomedicine. There are various computational methods for drug repurposing, one of which is using transcription signatures, especially single-cell RNA sequencing (scRNA-seq) data, which show us a clear and comprehensive view of the inside of the cell to compare the state of disease and health. METHODS In this study, we used 91,103 scRNA-seq samples from 29 patients with colorectal cancer (GSE144735 and GSE132465). First, differential gene expression (DGE) analysis was done using the ASAP website. Then we reached a list of drugs that can reverse the gene signature pattern from cancer to normal using the iLINCS website. Further, by searching various databases and articles, we found 12 drugs that have FDA approval, and so far, no one has reported them as a drug in the treatment of any cancer. Then, to evaluate the cytotoxicity and performance of these drugs, the MTT assay and real-time PCR were performed on two colorectal cancer cell lines (HT29 and HCT116). RESULTS According to our approach, 12 drugs were suggested for the treatment of colorectal cancer. Four drugs were selected for biological evaluation. The results of the cytotoxicity analysis of these drugs are as follows: tezacaftor (IC10 = 19 µM for HCT-116 and IC10 = 2 µM for HT-29), fenticonazole (IC10 = 17 µM for HCT-116 and IC10 = 7 µM for HT-29), bempedoic acid (IC10 = 78 µM for HCT-116 and IC10 = 65 µM for HT-29), and famciclovir (IC10 = 422 µM for HCT-116 and IC10 = 959 µM for HT-29). CONCLUSIONS Cost, time, and effectiveness are the main challenges in finding new drugs for diseases. Computational approaches such as transcriptional signature-based drug repurposing methods open new horizons to solve these challenges. In this study, tezacaftor, fenticonazole, and bempedoic acid can be introduced as promising drug candidates for the treatment of colorectal cancer. These drugs were evaluated in silico and in vitro, but it is necessary to evaluate them in vivo.
Collapse
Affiliation(s)
- Roohallah Mahdi-Esferizi
- Department of Medical Biotechnology, School of Advanced Technologies, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Zahra Shiasi
- Department of Medical Biotechnology, School of Advanced Technologies, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Razieh Heidari
- Department of Medical Biotechnology, School of Advanced Technologies, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Ali Najafi
- Molecular Biology Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Issa Mahmoudi
- Information Technology Department, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Fatemeh Elahian
- Department of Medical Biotechnology, School of Advanced Technologies, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Shahram Tahmasebian
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran.
| |
Collapse
|
17
|
Yang F, Akhtar MN, Zhang D, El-Mayta R, Shin J, Dorsey JF, Zhang L, Xu X, Guo W, Bagley SJ, Fuchs SY, Koumenis C, Lathia JD, Mitchell MJ, Gong Y, Fan Y. An immunosuppressive vascular niche drives macrophage polarization and immunotherapy resistance in glioblastoma. SCIENCE ADVANCES 2024; 10:eadj4678. [PMID: 38416830 PMCID: PMC10901371 DOI: 10.1126/sciadv.adj4678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Accepted: 01/25/2024] [Indexed: 03/01/2024]
Abstract
Cancer immunity is subjected to spatiotemporal regulation by leukocyte interaction with neoplastic and stromal cells, contributing to immune evasion and immunotherapy resistance. Here, we identify a distinct mesenchymal-like population of endothelial cells (ECs) that form an immunosuppressive vascular niche in glioblastoma (GBM). We reveal a spatially restricted, Twist1/SATB1-mediated sequential transcriptional activation mechanism, through which tumor ECs produce osteopontin to promote immunosuppressive macrophage (Mφ) phenotypes. Genetic or pharmacological ablation of Twist1 reverses Mφ-mediated immunosuppression and enhances T cell infiltration and activation, leading to reduced GBM growth and extended mouse survival, and sensitizing tumor to chimeric antigen receptor T immunotherapy. Thus, these findings uncover a spatially restricted mechanism controlling tumor immunity and suggest that targeting endothelial Twist1 may offer attractive opportunities for optimizing cancer immunotherapy.
Collapse
Affiliation(s)
- Fan Yang
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Md Naushad Akhtar
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Duo Zhang
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Rakan El-Mayta
- Department of Bioengineering, University of Pennsylvania School of Engineering and Applied Science, Philadelphia, PA 19104, USA
| | - Junyoung Shin
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jay F. Dorsey
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Lin Zhang
- Department of Obstetrics and Gynecology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Xiaowei Xu
- Department of Pathology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Wei Guo
- Department of Biology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Stephen J. Bagley
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Serge Y Fuchs
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Constantinos Koumenis
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Justin D. Lathia
- Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Michael J. Mitchell
- Department of Bioengineering, University of Pennsylvania School of Engineering and Applied Science, Philadelphia, PA 19104, USA
| | - Yanqing Gong
- Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Yi Fan
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA 19104, USA
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
18
|
Cai H, Ke ZB, Chen JY, Li XD, Zhu JM, Xue YT, Ruan ZT, Wang Z, Lin F, Zheng QS, Wei Y, Xue XY, Xu N. Ubiquitin-specific protease 5 promotes bladder cancer progression through stabilizing Twist1. Oncogene 2024; 43:703-713. [PMID: 38218898 DOI: 10.1038/s41388-023-02936-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 12/17/2023] [Accepted: 12/22/2023] [Indexed: 01/15/2024]
Abstract
Aberrant activation of the epithelial-mesenchymal transition (EMT) pathway drives the development of solid tumors, which is precisely regulated by core EMT-related transcription factors, including Twist1. However, the expression pattern and regulatory mechanism of Twist1 in the progression of bladder cancer is still unclear. In this study, we explore the role of Twist1 in the progression of bladder cancer. We discovered that the EMT regulon Twist1 protein, but not Twist1 mRNA, is overexpressed in bladder cancer samples using RT-qPCR, western blot and immunohistochemistry (IHC). Mechanistically, co-immunoprecipitation (Co-IP) coupled with liquid chromatography and tandem mass spectrometry identified USP5 as a binding partner of Twist1, and the binding of Twist1 to ubiquitin-specific protease 5 (USP5) stabilizes Twist through its deubiquitinase activity to activate the EMT. Further studies found that USP5 depletion reduces cell proliferation, invasion and the EMT in bladder cancer cells, and ectopic expression of Twist1 rescues the adverse effects of USP5 loss on cell invasion and the EMT. A xenograft tumor model was used to reconfirmed the inhibitor effect of silencing USP5 expression on tumorigenesis in vivo. In addition, USP5 protein levels are significantly elevated and positively associated with Twist1 levels in clinical bladder cancer samples. Collectively, our study revealed that USP5-Twist1 axis is a novel regulatory mechanism driving bladder cancer progression and that approaches targeting USP5 may become a promising cancer treatment strategy.
Collapse
Affiliation(s)
- Hai Cai
- Department of Urology, Urology Research Institute, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
- Department of Urology, National Region Medical center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China
| | - Zhi-Bin Ke
- Department of Urology, Urology Research Institute, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
- Department of Urology, National Region Medical center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China
| | - Jia-Yin Chen
- Department of Urology, Urology Research Institute, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
- Department of Urology, National Region Medical center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China
| | - Xiao-Dong Li
- Department of Urology, Urology Research Institute, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
- Department of Urology, National Region Medical center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China
| | - Jun-Ming Zhu
- Department of Urology, Urology Research Institute, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
- Department of Urology, National Region Medical center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China
| | - Yu-Ting Xue
- Department of Urology, Urology Research Institute, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
- Department of Urology, National Region Medical center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China
| | - Zhong-Tian Ruan
- Department of Urology, Urology Research Institute, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
- Department of Urology, National Region Medical center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China
| | - Zhen Wang
- Department of Urology, Urology Research Institute, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
- Department of Urology, National Region Medical center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China
| | - Fei Lin
- Department of Urology, Urology Research Institute, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
- Department of Urology, National Region Medical center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China
| | - Qing-Shui Zheng
- Department of Urology, Urology Research Institute, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
- Department of Urology, National Region Medical center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China
| | - Yong Wei
- Department of Urology, Urology Research Institute, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
- Department of Urology, National Region Medical center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China
| | - Xue-Yi Xue
- Department of Urology, Urology Research Institute, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China.
- Department of Urology, National Region Medical center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China.
- Fujian Key Laboratory of Precision Medicine for Cancer, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China.
| | - Ning Xu
- Department of Urology, Urology Research Institute, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China.
- Department of Urology, National Region Medical center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China.
- Fujian Key Laboratory of Precision Medicine for Cancer, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China.
| |
Collapse
|
19
|
Strippoli R, Niayesh-Mehr R, Adelipour M, Khosravi A, Cordani M, Zarrabi A, Allameh A. Contribution of Autophagy to Epithelial Mesenchymal Transition Induction during Cancer Progression. Cancers (Basel) 2024; 16:807. [PMID: 38398197 PMCID: PMC10886827 DOI: 10.3390/cancers16040807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 02/13/2024] [Accepted: 02/13/2024] [Indexed: 02/25/2024] Open
Abstract
Epithelial Mesenchymal Transition (EMT) is a dedifferentiation process implicated in many physio-pathological conditions including tumor transformation. EMT is regulated by several extracellular mediators and under certain conditions it can be reversible. Autophagy is a conserved catabolic process in which intracellular components such as protein/DNA aggregates and abnormal organelles are degraded in specific lysosomes. In cancer, autophagy plays a controversial role, acting in different conditions as both a tumor suppressor and a tumor-promoting mechanism. Experimental evidence shows that deep interrelations exist between EMT and autophagy-related pathways. Although this interplay has already been analyzed in previous studies, understanding mechanisms and the translational implications of autophagy/EMT need further study. The role of autophagy in EMT is not limited to morphological changes, but activation of autophagy could be important to DNA repair/damage system, cell adhesion molecules, and cell proliferation and differentiation processes. Based on this, both autophagy and EMT and related pathways are now considered as targets for cancer therapy. In this review article, the contribution of autophagy to EMT and progression of cancer is discussed. This article also describes the multiple connections between EMT and autophagy and their implication in cancer treatment.
Collapse
Affiliation(s)
- Raffaele Strippoli
- Department of Molecular Medicine, Sapienza University of Rome, 00161 Rome, Italy;
- National Institute for Infectious Diseases “Lazzaro Spallanzani”, I.R.C.C.S., 00149 Rome, Italy
| | - Reyhaneh Niayesh-Mehr
- Department of Clinical Biochemistry, Faculty of Medical Science, Tarbiat Modares University, Tehran P.O. Box 14115-331, Iran;
| | - Maryam Adelipour
- Department of Clinical Biochemistry, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz 61357-15794, Iran;
| | - Arezoo Khosravi
- Department of Genetics and Bioengineering, Faculty of Engineering and Natural Sciences, Istanbul Okan University, Istanbul 34959, Türkiye;
| | - Marco Cordani
- Department of Biochemistry and Molecular Biology, Faculty of Biological Sciences, Complutense University of Madrid, 28040 Madrid, Spain;
- Instituto de Investigaciones Sanitarias San Carlos (IdISSC), 28040 Madrid, Spain
| | - Ali Zarrabi
- Department of Biomedical Engineering, Faculty of Engineering and Natural Sciences, Istinye University, Istanbul 34396, Türkiye;
- Department of Research Analytics, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai 600077, India
| | - Abdolamir Allameh
- Department of Clinical Biochemistry, Faculty of Medical Science, Tarbiat Modares University, Tehran P.O. Box 14115-331, Iran;
| |
Collapse
|
20
|
Mukerjee N, Nag S, Bhattacharya B, Alexiou A, Mirgh D, Mukherjee D, Adhikari MD, Anand K, Muthusamy R, Gorai S, Thorat N. Clinical impact of epithelial–mesenchymal transition for cancer therapy. CLINICAL AND TRANSLATIONAL DISCOVERY 2024; 4. [DOI: 10.1002/ctd2.260] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Accepted: 12/26/2023] [Indexed: 01/03/2025]
Abstract
AbstractThe epithelial–mesenchymal transition (EMT) represents a pivotal frontier in oncology, playing a central role in the metastatic cascade of cancer—a leading global health challenge. This comprehensive review delves into the complexities of EMT, a process where cancer cells gain exceptional mobility, facilitating their invasion into distant organs and the establishment of secondary malignancies. We thoroughly examine the myriad of factors influencing EMT, encompassing transcription factors, signalling pathways, metabolic alterations, microRNAs, long non‐coding RNAs, epigenetic changes, exosomal interactions and the intricate dynamics of the tumour microenvironment. Particularly, the review emphasises the advanced stages of EMT, crucial for the development of highly aggressive cancer phenotypes. During this phase, cancer cells penetrate the vascular barrier and exploit the bloodstream to propagate life‐threatening metastases through the mesenchymal–epithelial transition. We also explore EMT's significant role in fostering tumour dormancy, senescence, the emergence of cancer stem cells and the formidable challenge of therapeutic resistance. Our review transcends a mere inventory of EMT‐inducing elements; it critically assesses the current state of EMT‐focused clinical trials, revealing both the hurdles and significant breakthroughs. Highlighting the potential of EMT research, we project its transformative impact on the future of cancer therapy. This exploration is aimed at paving the way towards an era of effectively managing this relentless disease, positioning EMT at the forefront of innovative cancer research strategies.
Collapse
Affiliation(s)
- Nobendu Mukerjee
- Department of Microbiology West Bengal State University, Barasat Kolkata India
| | - Sagnik Nag
- Department of Bio‐Sciences School of Biosciences & Technology Vellore Institute of Technology Vellore Tamil Nadu India
| | - Bikramjit Bhattacharya
- Department of Applied Microbiology School of Biosciences and Technology Vellore Institute of Technology Vellore Tamil Nadu India
| | - Athanasios Alexiou
- Department of Science and Engineering Novel Global Community Educational Foundation Hebersham New South Wales Australia
| | - Divya Mirgh
- Vaccine and Immunotherapy Canter Massachusetts General Hospital Boston Massachusetts USA
| | | | - Manab Deb Adhikari
- Department of Biotechnology University of North Bengal Darjeeling West Bengal India
| | - Krishnan Anand
- Department of Chemical Pathology School of Pathology Faculty of Health Sciences University of the Free State Bloemfontein South Africa
| | - Raman Muthusamy
- Center for Global Health Research Saveetha Medical College & Hospitals, Saveetha Institute of Medical and Technical Sciences Chennai Tamil Nadu India
| | | | - Nanasaheb Thorat
- Limerick Digital Cancer Research Centre and Department of Physics Bernal Institute University of Limerick, Castletroy Limerick Ireland
| |
Collapse
|
21
|
Li Z, Teng L, Pan Z, Yang Y, Zhu J, Wu X, Qian Y, Qian H, Bian Y, Chen Y, Chen W, Bi L. Identification of Comprehensive Biomarkers in Patients With Mismatch Repair-Deficient Colon Adenocarcinoma Based on Parallel Multiomics. J Transl Med 2024; 104:100306. [PMID: 38104864 DOI: 10.1016/j.labinv.2023.100306] [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: 04/24/2023] [Revised: 11/14/2023] [Accepted: 12/11/2023] [Indexed: 12/19/2023] Open
Abstract
Immunocheckpoint inhibitors have shown impressive efficacy in patients with colon cancer and other types of solid tumor that are mismatch repair-deficient (dMMR). Currently, PCR-capillary electrophoresis is one of the mainstream detection methods for dMMR, but its accuracy is still limited by germline mismatch repair (MMR) mutations, the functional redundancy of the MMR system, and abnormal methylation of MutL Homolog 1 promoter. Therefore, this study aimed to develop new biomarkers for dMMR based on artificial intelligence (AI) and pathologic images, which may help to improve the detection accuracy. To screen for the differential expression genes (DEGs) in dMMR patients and validate their diagnostic and prognostic efficiency, we used the expression profile data from the Cancer Genome Atlas (TCGA). The results showed that the expression of Immunoglobulin Lambda Joining 3 in dMMR patients was significantly downregulated and negatively correlated with the prognosis. Meanwhile, our diagnostic models based on pathologic image features showed good performance with area under the curves (AUCs) of 0.73, 0.86, and 0.81 in the training, test, and external validation sets (Jiangsu Traditional Chinese Medicine Hospital cohort). Based on gene expression and pathologic characteristics, we developed an effective prognosis model for dMMR patients through multiple Cox regression analysis (with AUC values of 0.88, 0.89, and 0.88 at 1-, 3-, and 5-year intervals, respectively). In conclusion, our results showed that Immunoglobulin Lambda Joining 3 and nucleus shape-related parameters (such as nuclear texture, nuclear eccentricity, nuclear size, and nuclear pixel intensity) were independent diagnostic and prognostic factors, suggesting that they could be used as new biomarkers for dMMR patients.
Collapse
Affiliation(s)
- Zhengjun Li
- College of Health Economics Management, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China; Jiangsu Provincial Engineering Research Center of TCM External Medication Development and Application, Nanjing, China
| | - Linxin Teng
- Jiangsu Provincial Engineering Research Center of TCM External Medication Development and Application, Nanjing, China; School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Zhiwei Pan
- School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Yang Yang
- School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Junlin Zhu
- School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Xiaobin Wu
- Affiliated Hospital, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Yunzhi Qian
- MPH Department of Nutrition, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Haihua Qian
- Affiliated Hospital, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Yaoyao Bian
- Jiangsu Provincial Engineering Research Center of TCM External Medication Development and Application, Nanjing, China
| | - Ying Chen
- College of Health Economics Management, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Weiping Chen
- School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China.
| | - Lei Bi
- Jiangsu Provincial Engineering Research Center of TCM External Medication Development and Application, Nanjing, China; School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China.
| |
Collapse
|
22
|
Meng W, Xiao H, Zhao R, Chen J, Wang Y, Mei P, Li H, Liao Y. METTL3 drives NSCLC metastasis by enhancing CYP19A1 translation and oestrogen synthesis. Cell Biosci 2024; 14:10. [PMID: 38238831 PMCID: PMC10795463 DOI: 10.1186/s13578-024-01194-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Accepted: 01/10/2024] [Indexed: 01/22/2024] Open
Abstract
BACKGROUND METTL3 plays a significant role as a catalytic enzyme in mediating N6-methyladenosine (m6A) modification, and its importance in tumour progression has been extensively studied in recent years. However, the precise involvement of METTL3 in the regulation of translation in non-small cell lung cancer (NSCLC) remains unclear. RESULTS Here we discovered by clinical investigation that METTL3 expression is correlated with NSCLC metastasis. Ablation of METTL3 in NSCLC cells inhibits invasion and metastasis in vitro and in vivo. Subsequently, through translatomics data mining and experimental validation, we demonstrated that METTL3 enhances the translation of aromatase (CYP19A1), a key enzyme in oestrogen synthesis, thereby promoting oestrogen production and mediating the invasion and metastasis of NSCLC. Mechanistically, METTL3 interacts with translation initiation factors and binds to CYP19A1 mRNA, thus enhancing the translation efficiency of CYP19A1 in an m6A-dependent manner. Pharmacological inhibition of METTL3 enzymatic activity or translation initiation factor eIF4E abolishes CYP19A1 protein synthesis. CONCLUSIONS Our findings indicate the crucial role of METTL3-mediated translation regulation in NSCLC and reveal the significance of METTL3/eIF4E/CYP19A1 signaling as a promising therapeutic target for anti-metastatic strategies against NSCLC.
Collapse
Affiliation(s)
- Wangyang Meng
- Department of Thoracic Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Han Xiao
- Department of Thoracic Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, China.
| | - Rong Zhao
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Jiaping Chen
- Department of Cardiothoracic Surgery, Third Affiliated Hospital of Kunming Medical University (Yunnan Cancer Hospital), Kunming, Yunnan, China
| | - Yangwei Wang
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Peiyuan Mei
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Hecheng Li
- Department of Thoracic Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Yongde Liao
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
23
|
Liu E, Qian X, He Y, Chen K. FUT4 promotes the progression of Cholangiocarcinoma by modulating epithelial-mesenchymal transition. Cell Cycle 2024; 23:218-231. [PMID: 38466946 PMCID: PMC11037297 DOI: 10.1080/15384101.2024.2318949] [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: 11/11/2023] [Accepted: 02/02/2024] [Indexed: 03/13/2024] Open
Abstract
Cholangiocarcinoma (CCA) is a common gastrointestinal malignancy characterized by a poor prognosis. Considering its prevalence, exploring its underlying molecular biological mechanisms is of paramount clinical importance. In this study, bioinformatics techniques were utilized to analyze CCA sample data obtained from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) databases. The analysis revealed a notable upregulation in FUT4 expression in CCA samples. To further investigate the functional implications of FUT4, in vivo and in vitro experiments were conducted, which demonstrated that FUT4 overexpression significantly enhances the proliferative and migratory capabilities of tumor cells. Subsequent sequencing analysis unveiled a correlation between FUT4 and epithelial-mesenchymal transition (EMT). Indeed, the pioneering discovery of elevated FUT4 expression in CCA was highlighted in this study. Further investigations into the function of FUT4 in CCA provided initial insights into its role in driving cancer progression via EMT. These findings present promising avenues for the diagnosis and treatment of CCA.[Figure: see text].
Collapse
Affiliation(s)
- Enchi Liu
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, P. R. China
| | - Xingwang Qian
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, P. R. China
| | - Yuan He
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, P. R. China
| | - Kunlun Chen
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, P. R. China
| |
Collapse
|
24
|
Peglion F, Etienne-Manneville S. Cell polarity changes in cancer initiation and progression. J Cell Biol 2024; 223:e202308069. [PMID: 38091012 PMCID: PMC10720656 DOI: 10.1083/jcb.202308069] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Revised: 11/20/2023] [Accepted: 11/21/2023] [Indexed: 12/17/2023] Open
Abstract
Cell polarity, which consists of the morphological, structural, and functional organization of cells along a defined axis, is a feature of healthy cells and tissues. In contrast, abnormal polarity is a hallmark of cancer cells. At the molecular level, key evolutionarily conserved proteins that control polarity establishment and maintenance in various contexts are frequently altered in cancer, but the relevance of these molecular alterations in the oncogenic processes is not always clear. Here, we summarize the recent findings, shedding new light on the involvement of polarity players in cancer development, and discuss the possibility of harnessing cell polarity changes to better predict, diagnose, and cure cancers.
Collapse
Affiliation(s)
- Florent Peglion
- Cell Polarity, Migration and Cancer Unit, Université de Paris, UMR3691 CNRS, Equipe Labellisée Ligue 2023, Institut Pasteur, Paris, France
| | - Sandrine Etienne-Manneville
- Cell Polarity, Migration and Cancer Unit, Université de Paris, UMR3691 CNRS, Equipe Labellisée Ligue 2023, Institut Pasteur, Paris, France
| |
Collapse
|
25
|
Chai XX, Liu J, Yu TY, Zhang G, Sun WJ, Zhou Y, Ren L, Cao HL, Yin DC, Zhang CY. Recent progress of mechanosensitive mechanism on breast cancer. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2023; 185:1-16. [PMID: 37793504 DOI: 10.1016/j.pbiomolbio.2023.09.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 08/10/2023] [Accepted: 09/25/2023] [Indexed: 10/06/2023]
Abstract
The mechanical environment is important for tumorigenesis and progression. Tumor cells can sense mechanical signals by mechanosensitive receptors, and these mechanical signals can be converted to biochemical signals to regulate cell behaviors, such as cell differentiation, proliferation, migration, apoptosis, and drug resistance. Here, we summarized the effects of the mechanical microenvironment on breast cancer cell activity, and mechanotransduction mechanism from cellular microenvironment to cell membrane, and finally to the nucleus, and also relative mechanosensitive proteins, ion channels, and signaling pathways were elaborated, therefore the mechanical signal could be transduced to biochemical or molecular signal. Meanwhile, the mechanical models commonly used for biomechanics study in vitro and some quantitative descriptions were listed. It provided an essential theoretical basis for the occurrence and development of mechanosensitive breast cancer, and also some potential drug targets were proposed to treat such disease.
Collapse
Affiliation(s)
- Xiao-Xia Chai
- Institute for Special Environmental Biophysics, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, Shaanxi, PR China
| | - Jie Liu
- Institute for Special Environmental Biophysics, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, Shaanxi, PR China
| | - Tong-Yao Yu
- Institute for Special Environmental Biophysics, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, Shaanxi, PR China
| | - Ge Zhang
- Institute for Special Environmental Biophysics, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, Shaanxi, PR China
| | - Wen-Jun Sun
- Institute for Special Environmental Biophysics, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, Shaanxi, PR China
| | - Yan Zhou
- Institute for Special Environmental Biophysics, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, Shaanxi, PR China
| | - Li Ren
- Institute for Special Environmental Biophysics, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, Shaanxi, PR China; Key Laboratory of Flexible Electronics of Zhejiang Province, Ningbo Institute of Northwestern Polytechnical University, Ningbo, 315103, Zhejiang, PR China
| | - Hui-Ling Cao
- Xi'an Key Laboratory of Basic and Translation of Cardiovascular Metabolic Disease, School of Pharmacy, Xi'an Medical University, Xi'an, 710021, Shaanxi, PR China.
| | - Da-Chuan Yin
- Institute for Special Environmental Biophysics, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, Shaanxi, PR China.
| | - Chen-Yan Zhang
- Institute for Special Environmental Biophysics, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, Shaanxi, PR China.
| |
Collapse
|
26
|
Yu X, He T, Tong Z, Liao L, Huang S, Fakhouri WD, Edwards DP, Xu J. Molecular mechanisms of TWIST1-regulated transcription in EMT and cancer metastasis. EMBO Rep 2023; 24:e56902. [PMID: 37680145 PMCID: PMC10626429 DOI: 10.15252/embr.202356902] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 08/23/2023] [Accepted: 08/25/2023] [Indexed: 09/09/2023] Open
Abstract
TWIST1 induces epithelial-to-mesenchymal transition (EMT) to drive cancer metastasis. It is yet unclear what determines TWIST1 functions to activate or repress transcription. We found that the TWIST1 N-terminus antagonizes TWIST1-regulated gene expression, cancer growth and metastasis. TWIST1 interacts with both the NuRD complex and the NuA4/TIP60 complex (TIP60-Com) via its N-terminus. Non-acetylated TWIST1-K73/76 selectively interacts with and recruits NuRD to repress epithelial target gene transcription. Diacetylated TWIST1-acK73/76 binds BRD8, a component of TIP60-Com that also binds histone H4-acK5/8, to recruit TIP60-Com to activate mesenchymal target genes and MYC. Knockdown of BRD8 abolishes TWIST1 and TIP60-Com interaction and TIP60-Com recruitment to TWIST1-activated genes, resulting in decreasing TWIST1-activated target gene expression and cancer metastasis. Both TWIST1/NuRD and TWIST1/TIP60-Com complexes are required for TWIST1 to promote EMT, proliferation, and metastasis at full capacity. Therefore, the diacetylation status of TWIST1-K73/76 dictates whether TWIST1 interacts either with NuRD to repress epithelial genes, or with TIP60-Com to activate mesenchymal genes and MYC. Since BRD8 is essential for TWIST1-acK73/76 and TIP60-Com interaction, targeting BRD8 could be a means to inhibit TWIST1-activated gene expression.
Collapse
Affiliation(s)
- Xiaobin Yu
- Department of Molecular and Cellular BiologyBaylor College of MedicineHoustonTXUSA
| | - Tao He
- Department of Molecular and Cellular BiologyBaylor College of MedicineHoustonTXUSA
- Present address:
Institute for Cancer MedicineSouthwest Medical UniversitySichuanChina
| | - Zhangwei Tong
- Department of Molecular and Cellular BiologyBaylor College of MedicineHoustonTXUSA
| | - Lan Liao
- Department of Molecular and Cellular BiologyBaylor College of MedicineHoustonTXUSA
- Dan L. Duncan Comprehensive Cancer CenterBaylor College of MedicineHoustonTXUSA
| | - Shixia Huang
- Department of Molecular and Cellular BiologyBaylor College of MedicineHoustonTXUSA
- Dan L. Duncan Comprehensive Cancer CenterBaylor College of MedicineHoustonTXUSA
| | - Walid D Fakhouri
- Department of Diagnostic and Biomedical Sciences, Center for Craniofacial Research, School of DentistryUniversity of Texas Health Science Center at HoustonHoustonTXUSA
| | - Dean P Edwards
- Department of Molecular and Cellular BiologyBaylor College of MedicineHoustonTXUSA
- Dan L. Duncan Comprehensive Cancer CenterBaylor College of MedicineHoustonTXUSA
| | - Jianming Xu
- Department of Molecular and Cellular BiologyBaylor College of MedicineHoustonTXUSA
- Dan L. Duncan Comprehensive Cancer CenterBaylor College of MedicineHoustonTXUSA
| |
Collapse
|
27
|
Qiang L, Zhao B, Ming M, Wang N, He TC, Hwang S, Thorburn A, He YY. Autophagy regulates tumor growth and metastasis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.31.564991. [PMID: 37961427 PMCID: PMC10635024 DOI: 10.1101/2023.10.31.564991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
The role of autophagy in tumorigenesis and tumor metastasis remains poorly understood. Here we show that inhibition of autophagy stabilizes the transcription factor Twist1 through Sequestosome-1 (SQSTM1, also known as p62) and thus increases cell proliferation, migration, and epithelial-mesenchymal transition (EMT) in tumor development and metastasis. Inhibition of autophagy or p62 overexpression blocks Twist1 protein degradation in the proteasomes, while p62 inhibition enhances it. SQSTM1/p62 interacts with Twist1 via the UBA domain of p62, in a Twist1-ubiquitination-dependent manner. Lysine 175 in Twist1 is critical for Twist1 ubiquitination, degradation, and SQSTM1/p62 interaction. For squamous skin cancer and melanoma cells that express Twist1, SQSTM1/p62 increases tumor growth and metastasis in mice. Together, our results identified Twist1 as a key downstream protein for autophagy and suggest a critical role of the autophagy/p62/Twist1 axis in cancer development and metastasis.
Collapse
Affiliation(s)
- Lei Qiang
- Department of Medicine, Section of Dermatology, University of Chicago, Chicago, IL, USA
| | - Baozhong Zhao
- Department of Medicine, Section of Dermatology, University of Chicago, Chicago, IL, USA
| | - Mei Ming
- Department of Medicine, Section of Dermatology, University of Chicago, Chicago, IL, USA
| | - Ning Wang
- Department of Orthopaedic Surgery & Rehabilitation Medicine, University of Chicago, Chicago, IL, USA
| | - Tong-Chuan He
- Department of Orthopaedic Surgery & Rehabilitation Medicine, University of Chicago, Chicago, IL, USA
| | - Seungmin Hwang
- Department of Pathology, University of Chicago, Chicago, IL, USA
| | - Andrew Thorburn
- Department of Pharmacology, University of Colorado at Denver and Health Sciences Center, Aurora, Colorado, USA
| | - Yu-Ying He
- Department of Medicine, Section of Dermatology, University of Chicago, Chicago, IL, USA
| |
Collapse
|
28
|
Bukhari M, Patel N, Fontana R, Santiago-Medina M, Jiang Y, Li D, Pestonjamasp K, Christiansen VJ, Jackson KW, McKee PA, Yang J. Fibroblast activation protein drives tumor metastasis via a protease-independent role in invadopodia stabilization. Cell Rep 2023; 42:113302. [PMID: 37862167 PMCID: PMC10742343 DOI: 10.1016/j.celrep.2023.113302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 08/09/2023] [Accepted: 10/03/2023] [Indexed: 10/22/2023] Open
Abstract
During metastasis, tumor cells invade through the basement membrane and intravasate into blood vessels and then extravasate into distant organs to establish metastases. Here, we report a critical role of a transmembrane serine protease fibroblast activation protein (FAP) in tumor metastasis. Expression of FAP and TWIST1, a metastasis driver, is significantly correlated in several types of human carcinomas, and FAP is required for TWIST1-induced breast cancer metastasis to the lung. Mechanistically, FAP is localized at invadopodia and required for invadopodia-mediated extracellular matrix degradation independent of its proteolytic activity. Live cell imaging shows that association of invadopodia precursors with FAP at the cell membrane promotes the stabilization and growth of invadopodia precursors into mature invadopodia. Together, our study identified FAP as a functional target of TWIST1 in driving tumor metastasis via promoting invadopodia-mediated matrix degradation and uncovered a proteolytic activity-independent role of FAP in stabilizing invadopodia precursors for maturation.
Collapse
Affiliation(s)
- Maurish Bukhari
- Department of Pharmacology, University of California, San Diego, School of Medicine, La Jolla, CA 92093, USA; Moores Cancer Center, University of California, San Diego, School of Medicine, La Jolla, CA 92093, USA
| | - Navneeta Patel
- Department of Pharmacology, University of California, San Diego, School of Medicine, La Jolla, CA 92093, USA; Moores Cancer Center, University of California, San Diego, School of Medicine, La Jolla, CA 92093, USA
| | - Rosa Fontana
- Department of Pharmacology, University of California, San Diego, School of Medicine, La Jolla, CA 92093, USA; Moores Cancer Center, University of California, San Diego, School of Medicine, La Jolla, CA 92093, USA
| | - Miguel Santiago-Medina
- Department of Pharmacology, University of California, San Diego, School of Medicine, La Jolla, CA 92093, USA; Moores Cancer Center, University of California, San Diego, School of Medicine, La Jolla, CA 92093, USA
| | - Yike Jiang
- Department of Pharmacology, University of California, San Diego, School of Medicine, La Jolla, CA 92093, USA; Moores Cancer Center, University of California, San Diego, School of Medicine, La Jolla, CA 92093, USA
| | - Dongmei Li
- Department of Pharmacology, University of California, San Diego, School of Medicine, La Jolla, CA 92093, USA; Moores Cancer Center, University of California, San Diego, School of Medicine, La Jolla, CA 92093, USA
| | - Kersi Pestonjamasp
- Moores Cancer Center, University of California, San Diego, School of Medicine, La Jolla, CA 92093, USA
| | - Victoria J Christiansen
- William K. Warren Medical Research Center, Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Kenneth W Jackson
- William K. Warren Medical Research Center, Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Patrick A McKee
- William K. Warren Medical Research Center, Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Jing Yang
- Department of Pharmacology, University of California, San Diego, School of Medicine, La Jolla, CA 92093, USA; Moores Cancer Center, University of California, San Diego, School of Medicine, La Jolla, CA 92093, USA; Department of Pediatrics, University of California, San Diego, School of Medicine, La Jolla, CA 92093, USA.
| |
Collapse
|
29
|
Xu R, Yin P, Wei J, Ding Q. The role of matrix stiffness in breast cancer progression: a review. Front Oncol 2023; 13:1284926. [PMID: 37916166 PMCID: PMC10616305 DOI: 10.3389/fonc.2023.1284926] [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: 08/29/2023] [Accepted: 10/04/2023] [Indexed: 11/03/2023] Open
Abstract
The significance of matrix stiffness in cancer development has been investigated in recent years. The gradual elastic force the extracellular matrix imparts to cells, known as matrix stiffness, is one of the most important types of mechanical stimulation. Increased matrix stiffness alters the biological activity of cells, which promotes the growth of numerous malignancies, including breast cancer. Comprehensive studies have demonstrated that increasing matrix stiffness activates molecular signaling pathways that are closely linked to breast cancer progression. There are many articles exploring the relationship between mechanism hardness and breast cancer, so we wanted to provide a systematic summary of recent research advances. In this review, we briefly introduce the mechanism of matrix stiffness in breast cancer, elaborate on the effect of extracellular matrix stiffness on breast cancer biological behavior and signaling pathways, and finally, we will talk about breast cancer treatment that focuses on matrix stiffness.
Collapse
Affiliation(s)
- Ruoxi Xu
- Department of Pharmacy, The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing, China
- Jiangsu Breast Disease Center, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China
| | - Peng Yin
- Jiangsu Breast Disease Center, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China
| | - Jifu Wei
- Department of Pharmacy, The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing, China
| | - Qiang Ding
- Jiangsu Breast Disease Center, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China
| |
Collapse
|
30
|
Huang L, Xing Y, Ning X, Yu Z, Bai X, Liu L, Sun S. Roles of Twist1 in lipid and glucose metabolism. Cell Commun Signal 2023; 21:270. [PMID: 37784111 PMCID: PMC10544448 DOI: 10.1186/s12964-023-01262-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Accepted: 08/09/2023] [Indexed: 10/04/2023] Open
Abstract
The abnormal lipid and glucose metabolisms are linked to the metabolic disorders, tumorigenesis, and fibrotic diseases, which attracts the increasing attention to find out the key molecules involved in the lipid and glucose metabolism as the possible therapeutic targets on these diseases. A transcriptional factor Twist1 has been associated with not only the embryonic development, cancer, and fibrotic diseases, but also the regulation of lipid and glucose metabolism. In this review, we will discuss the roles and mechanisms of Twist1 in the obesity-associated white adipose tissue inflammation and insulin resistance, brown adipose tissue metabolism, fatty acid oxidation, and glucose metabolism in skeletal muscle to provide a rational perspective to consider Twist1 as a potential treatment target in clinic. Video Abstract.
Collapse
Affiliation(s)
- Liuyifei Huang
- Department of Nephrology, Xijing Hospital, The Fourth Military Medical University, Changle Road, No. 127 Changle West Road, Xi'an, Shaanxi, China
| | - Yan Xing
- Department of Nephrology, Xijing Hospital, The Fourth Military Medical University, Changle Road, No. 127 Changle West Road, Xi'an, Shaanxi, China
| | - Xiaoxuan Ning
- Department of Geriatrics, Xijing Hospital, The Fourth Military Medical University, Changle Road, No. 127 Changle West Road, Xi'an, Shaanxi, China
| | - Zhixiang Yu
- Department of Nephrology, Xijing Hospital, The Fourth Military Medical University, Changle Road, No. 127 Changle West Road, Xi'an, Shaanxi, China
| | - Xiao Bai
- Department of Nephrology, Xijing Hospital, The Fourth Military Medical University, Changle Road, No. 127 Changle West Road, Xi'an, Shaanxi, China
| | - Limin Liu
- School of Medicine, Northwest University, 229 Taibai North Road, Xi'an, 710032, Shaanxi, China.
| | - Shiren Sun
- Department of Nephrology, Xijing Hospital, The Fourth Military Medical University, Changle Road, No. 127 Changle West Road, Xi'an, Shaanxi, China.
| |
Collapse
|
31
|
Khan SU, Fatima K, Malik F, Kalkavan H, Wani A. Cancer metastasis: Molecular mechanisms and clinical perspectives. Pharmacol Ther 2023; 250:108522. [PMID: 37661054 DOI: 10.1016/j.pharmthera.2023.108522] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 08/22/2023] [Accepted: 08/29/2023] [Indexed: 09/05/2023]
Abstract
Metastatic progression combined with non-responsiveness towards systemic therapy often shapes the course of disease for cancer patients and commonly determines its lethal outcome. The complex molecular events that promote metastasis are a combination of both, the acquired pro-metastatic properties of cancer cells and a metastasis-permissive or -supportive tumor micro-environment (TME). Yet, dissemination is a challenging process for cancer cells that requires a series of events to enable cancer cell survival and growth. Metastatic cancer cells have to initially detach themselves from primary tumors, overcome the challenges of their intravasal journey and colonize distant sites that are suited for their metastases. The implicated obstacles including anoikis and immune surveillance, can be overcome by intricate intra- and extracellular signaling pathways, which we will summarize and discuss in this review. Further, emerging modulators of metastasis, like the immune-microenvironment, microbiome, sublethal cell death engagement, or the nervous system will be integrated into the existing working model of metastasis.
Collapse
Affiliation(s)
- Sameer Ullah Khan
- The University of Texas MD Anderson Cancer Center, Division of Genitourinary Medical Oncology, Holcombe Blvd, Houston, TX 77030, USA; Division of Cancer Pharmacology, CSIR-Indian Institute of Integrative Medicine, Jammu and Kashmir, India
| | - Kaneez Fatima
- Division of Cancer Pharmacology, CSIR-Indian Institute of Integrative Medicine, Jammu and Kashmir, India; Academy of Scientific and Innovative Research (ASIR), Ghaziabad 201002, India
| | - Fayaz Malik
- Division of Cancer Pharmacology, CSIR-Indian Institute of Integrative Medicine, Jammu and Kashmir, India; Academy of Scientific and Innovative Research (ASIR), Ghaziabad 201002, India.
| | - Halime Kalkavan
- Department of Medical Oncology, West German Cancer Center, University Hospital Essen, Essen, Germany; German Cancer Consortium (DKTK), Partner Site University Hospital Essen, Essen, Germany.
| | - Abubakar Wani
- St. Jude Children's Research Hospital, 262 Danny Thomas Pl, Memphis, TN 38105, United States.
| |
Collapse
|
32
|
Huang Q, Zhang R, Xia Y, Shen J, Dong H, Li X, Tao D, Xie D, Liu L. DAB2IP suppresses invadopodia formation through destabilizing ALK by interacting with USP10 in breast cancer. iScience 2023; 26:107606. [PMID: 37664607 PMCID: PMC10470318 DOI: 10.1016/j.isci.2023.107606] [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: 02/15/2023] [Revised: 05/26/2023] [Accepted: 08/08/2023] [Indexed: 09/05/2023] Open
Abstract
Invadopodia, being actin-rich membrane protrusions, play a vital role in tumor cell invasion and metastasis. Our previous studies have revealed some functions of the DOC-2/DAB2 interacting protein (DAB2IP) as a tumor suppressor. Nevertheless, the specific role and mechanism of DAB2IP in invadopodia formation remain unclear. Here, we find that DAB2IP effectively suppresses invadopodia formation and metastasis in breast cancer, both in vitro and in vivo. Additionally, DAB2IP could downregulate anaplastic lymphoma kinase (ALK), resulting in the inhibition of tyrosine phosphorylation of Cortactin and the prevention of invadopodia formation. DAB2IP competitively antagonizes the interaction between the deubiquitinating enzyme Ubiquitin-specific peptidase 10 (USP10) and ALK, leading to a decrease in the abundance of ALK protein. In summary, DAB2IP impairs the stability of ALK through USP10-dependent deubiquitination, suppressing Cortactin phosphorylation, thereby inhibiting invadopodia formation and metastasis of breast cancer cells. Furthermore, this study suggests a potential therapeutic strategy for breast cancer treatment.
Collapse
Affiliation(s)
- Qingwen Huang
- Molecular Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, P.R. China
- Department of GI Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, P.R. China
| | - Rui Zhang
- Molecular Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, P.R. China
- Department of GI Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, P.R. China
| | - Yun Xia
- Department of Breast and Thyroid Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, P.R. China
| | - Jie Shen
- Molecular Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, P.R. China
- Department of GI Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, P.R. China
| | - Hongliang Dong
- Molecular Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, P.R. China
- Department of GI Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, P.R. China
| | - Xiaolan Li
- Molecular Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, P.R. China
| | - Deding Tao
- Department of GI Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, P.R. China
| | - Daxing Xie
- Molecular Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, P.R. China
- Department of GI Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, P.R. China
| | - Liang Liu
- Molecular Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, P.R. China
- Department of GI Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, P.R. China
| |
Collapse
|
33
|
Khatoon F, Ali S, Kumar V, Elasbali AM, Alhassan HH, Alharethi SH, Islam A, Hassan MI. Pharmacological features, health benefits and clinical implications of honokiol. J Biomol Struct Dyn 2023; 41:7511-7533. [PMID: 36093963 DOI: 10.1080/07391102.2022.2120541] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Accepted: 08/29/2022] [Indexed: 10/14/2022]
Abstract
Honokiol (HNK) is a natural polyphenolic compound extracted from the bark and leaves of Magnolia grandiflora. It has been traditionally used as a medicinal compound to treat inflammatory diseases. HNK possesses numerous health benefits with a minimal level of toxicity. It can cross the blood-brain barrier and blood-cerebrospinal fluid, thus having significant bioavailability in the neurological tissues. HNK is a promising bioactive compound possesses neuroprotective, antimicrobial, anti-tumorigenic, anti-spasmodic, antidepressant, analgesic, and antithrombotic features . HNK can prevent the growth of several cancer types and haematological malignancies. Recent studies suggested its role in COVID-19 therapy. It binds effectively with several molecular targets, including apoptotic factors, chemokines, transcription factors, cell surface adhesion molecules, and kinases. HNK has excellent pharmacological features and a wide range of chemotherapeutic effects, and thus, researchers have increased interest in improving the therapeutic implications of HNK to the clinic as a novel agent. This review focused on the therapeutic implications of HNK, highlighting clinical and pharmacological features and the underlying mechanism of action.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Fatima Khatoon
- Amity Institute of Neuropsychology & Neurosciences, Amity University, Noida, India
| | - Sabeeha Ali
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India
| | - Vijay Kumar
- Amity Institute of Neuropsychology & Neurosciences, Amity University, Noida, India
| | - Abdelbaset Mohamed Elasbali
- Department of Clinical Laboratory Science, College of Applied Medical Sciences-Qurayyat, Jouf University, Saudi Arabia
| | - Hassan H Alhassan
- Department of Clinical Laboratory Science, College of Applied Medical Sciences-Qurayyat, Jouf University, Saudi Arabia
| | - Salem Hussain Alharethi
- Department of Biological Science, College of Arts and Science, Najran University, Najran, Saudia Arabia
| | - Asimul Islam
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India
| | - Md Imtaiyaz Hassan
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India
| |
Collapse
|
34
|
Luo JQ, Yang TW, Wu J, Lai HH, Zou LB, Chen WB, Zhou XM, Lv DJ, Cen SR, Long ZN, Mao YY, Zheng PX, Su XH, Xian ZY, Shu FP, Mao XM. Exosomal PGAM1 promotes prostate cancer angiogenesis and metastasis by interacting with ACTG1. Cell Death Dis 2023; 14:502. [PMID: 37542027 PMCID: PMC10403531 DOI: 10.1038/s41419-023-06007-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Revised: 06/15/2023] [Accepted: 07/17/2023] [Indexed: 08/06/2023]
Abstract
Tumor-derived exosomes and their contents promote cancer metastasis. Phosphoglycerate mutase 1 (PGAM1) is involved in various cancer-related processes. Nevertheless, the underlying mechanism of exosomal PGAM1 in prostate cancer (PCa) metastasis remains unclear. In this study, we performed in vitro and in vivo to determine the functions of exosomal PGAM1 in the angiogenesis of patients with metastatic PCa. We performed Glutathione-S-transferase pulldown, co-immunoprecipitation, western blotting and gelatin degradation assays to determine the pathway mediating the effect of exosomal PGAM1 in PCa. Our results revealed a significant increase in exosomal PGAM1 levels in the plasma of patients with metastatic PCa compared to patients with non-metastatic PCa. Furthermore, PGAM1 was a key factor initiating PCa cell metastasis by promoting invadopodia formation and could be conveyed by exosomes from PCa cells to human umbilical vein endothelial cells (HUVECs). In addition, exosomal PGAM1 could bind to γ-actin (ACTG1), which promotes podosome formation and neovascular sprouting in HUVECs. In vivo results revealed exosomal PGAM1 enhanced lung metastasis in nude mice injected with PCa cells via the tail vein. In summary, exosomal PGAM1 promotes angiogenesis and could be used as a liquid biopsy marker for PCa metastasis.
Collapse
Affiliation(s)
- Jun-Qi Luo
- Department of Urology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Tao-Wei Yang
- Department of Urology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Jun Wu
- Department of Urology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Hou-Hua Lai
- Department of Urology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Li-Bin Zou
- Department of Urology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Wen-Bin Chen
- Department of Urology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Xu-Min Zhou
- Department of Urology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Dao-Jun Lv
- Department of Urology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Sheng-Ren Cen
- Department of Urology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Zi-Ning Long
- Department of Urology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Yi-You Mao
- Department of Urology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Peng-Xiang Zheng
- Department of Urology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Xiao-Hong Su
- Department of Urology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Zhi-Yong Xian
- Department of Urology, Guangdong Provincial People's Hospital's Nanhai Hospital, 23 Pingzhouxiadong Road, Foshan, 528251, China.
| | - Fang-Peng Shu
- Department of Urology, Guangzhou Women and Children's Medical Center, National Children's Medical Center for South Central Region, Guangzhou Medical University, Guangzhou, Guangdong, China.
| | - Xiang-Ming Mao
- Department of Urology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China.
| |
Collapse
|
35
|
Liu X, Li J, Yang X, Li X, Kong J, Qi D, Zhang F, Sun B, Liu Y, Liu T. Carcinoma-associated fibroblast-derived lysyl oxidase-rich extracellular vesicles mediate collagen crosslinking and promote epithelial-mesenchymal transition via p-FAK/p-paxillin/YAP signaling. Int J Oral Sci 2023; 15:32. [PMID: 37532712 PMCID: PMC10397209 DOI: 10.1038/s41368-023-00236-1] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 07/05/2023] [Accepted: 07/05/2023] [Indexed: 08/04/2023] Open
Abstract
Carcinoma-associated fibroblasts (CAFs) are the main cellular components of the tumor microenvironment and promote cancer progression by modifying the extracellular matrix (ECM). The tumor-associated ECM is characterized by collagen crosslinking catalyzed by lysyl oxidase (LOX). Small extracellular vesicles (sEVs) mediate cell-cell communication. However, the interactions between sEVs and the ECM remain unclear. Here, we demonstrated that sEVs released from oral squamous cell carcinoma (OSCC)-derived CAFs induce collagen crosslinking, thereby promoting epithelial-mesenchymal transition (EMT). CAF sEVs preferably bound to the ECM rather than being taken up by fibroblasts and induced collagen crosslinking, and a LOX inhibitor or blocking antibody suppressed this effect. Active LOX (αLOX), but not the LOX precursor, was enriched in CAF sEVs and interacted with periostin, fibronectin, and bone morphogenetic protein-1 on the surface of sEVs. CAF sEV-associated integrin α2β1 mediated the binding of CAF sEVs to collagen I, and blocking integrin α2β1 inhibited collagen crosslinking by interfering with CAF sEV binding to collagen I. CAF sEV-induced collagen crosslinking promoted the EMT of OSCC through FAK/paxillin/YAP pathway. Taken together, these findings reveal a novel role of CAF sEVs in tumor ECM remodeling, suggesting a critical mechanism for CAF-induced EMT of cancer cells.
Collapse
Affiliation(s)
- Xue Liu
- Department of Oral Pathology, Shanghai Stomatological Hospital & School of Stomatology, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Craniomaxillofacial Development and Diseases, Fudan University, Shanghai, China
- Department of Orthodontics, Shanghai Stomatological Hospital & School of Stomatology, Fudan University, Shanghai, China
- School of Stomatology, Dalian Medical University, Dalian, China
| | - Jiao Li
- Department of Oral Pathology, Shanghai Stomatological Hospital & School of Stomatology, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Craniomaxillofacial Development and Diseases, Fudan University, Shanghai, China
- Department of Orthodontics, Shanghai Stomatological Hospital & School of Stomatology, Fudan University, Shanghai, China
- School of Stomatology, Dalian Medical University, Dalian, China
| | - Xuesong Yang
- Department of Biochemistry and Molecular Biology, Liaoning Provincial Core Lab of Glycobiology and Glycoengineering, Dalian Medical University, Dalian, China
| | - Xiaojie Li
- School of Stomatology, Dalian Medical University, Dalian, China
| | - Jing Kong
- School of Stomatology, Dalian Medical University, Dalian, China
| | - Dongyuan Qi
- Department of Oral Surgery, the First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Fuyin Zhang
- Department of Oral Surgery, the Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Bo Sun
- Department of Oral Surgery, the Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Yuehua Liu
- Shanghai Key Laboratory of Craniomaxillofacial Development and Diseases, Fudan University, Shanghai, China.
- Department of Orthodontics, Shanghai Stomatological Hospital & School of Stomatology, Fudan University, Shanghai, China.
| | - Tingjiao Liu
- Department of Oral Pathology, Shanghai Stomatological Hospital & School of Stomatology, Fudan University, Shanghai, China.
- Shanghai Key Laboratory of Craniomaxillofacial Development and Diseases, Fudan University, Shanghai, China.
| |
Collapse
|
36
|
Toriumi K, Onodera Y, Takehara T, Mori T, Hasei J, Shigi K, Iwawaki N, Ozaki T, Akagi M, Nakanishi M, Teramura T. LRRC15 expression indicates high level of stemness regulated by TWIST1 in mesenchymal stem cells. iScience 2023; 26:106946. [PMID: 37534184 PMCID: PMC10391581 DOI: 10.1016/j.isci.2023.106946] [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: 12/25/2022] [Revised: 04/09/2023] [Accepted: 05/19/2023] [Indexed: 08/04/2023] Open
Abstract
Mesenchymal stem cells (MSCs) are used as a major source for cell therapy, and its application is expanding in various diseases. On the other hand, reliable method to evaluate quality and therapeutic properties of MSC is limited. In this study, we focused on TWIST1 that is a transcription factor regulating stemness of MSCs and found that the transmembrane protein LRRC15 tightly correlated with the expression of TWIST1 and useful to expect TWIST1-regulated stemness of MSCs. The LRRC15-positive MSC populations in human and mouse bone marrow tissues highly expressed stemness-associated transcription factors and therapeutic cytokines, and showed better therapeutic effect in bleomycin-induced pulmonary fibrosis model mice. This study provides evidence for the important role of TWIST1 in the MSC stemness, and for the utility of the LRRC15 protein as a marker to estimate stem cell quality in MSCs before cell transplantation.
Collapse
Affiliation(s)
- Kensuke Toriumi
- Department of Orthopedic Surgery, Kindai University Faculty of Medicine, Osaka-sayama, Osaka, Japan
| | - Yuta Onodera
- Institute of Advanced Clinical Medicine, Kindai University Hospital, Osaka-sayama, Osaka, Japan
| | - Toshiyuki Takehara
- Institute of Advanced Clinical Medicine, Kindai University Hospital, Osaka-sayama, Osaka, Japan
| | - Tatsufumi Mori
- Life Science Institute, Kindai University, Osaka-sayama, Osaka, Japan
| | - Joe Hasei
- Department of Orthopedic Surgery, Okayama University Faculty of Medicine, Okayama, Okayama, Japan
| | - Kanae Shigi
- Institute of Advanced Clinical Medicine, Kindai University Hospital, Osaka-sayama, Osaka, Japan
| | - Natsumi Iwawaki
- Institute of Advanced Clinical Medicine, Kindai University Hospital, Osaka-sayama, Osaka, Japan
| | - Toshifumi Ozaki
- Department of Orthopedic Surgery, Okayama University Faculty of Medicine, Okayama, Okayama, Japan
| | - Masao Akagi
- Department of Orthopedic Surgery, Kindai University Faculty of Medicine, Osaka-sayama, Osaka, Japan
| | | | - Takeshi Teramura
- Institute of Advanced Clinical Medicine, Kindai University Hospital, Osaka-sayama, Osaka, Japan
| |
Collapse
|
37
|
Zhang J, Zhang Z, Huang Z, Li M, Yang F, Wu Z, Guo Q, Mei X, Lu B, Wang C, Wang Z, Ji L. Isotoosendanin exerts inhibition on triple-negative breast cancer through abrogating TGF- β-induced epithelial-mesenchymal transition via directly targeting TGF βR1. Acta Pharm Sin B 2023; 13:2990-3007. [PMID: 37521871 PMCID: PMC10372922 DOI: 10.1016/j.apsb.2023.05.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 03/07/2023] [Accepted: 03/14/2023] [Indexed: 08/01/2023] Open
Abstract
As the most aggressive breast cancer, triple-negative breast cancer (TNBC) is still incurable and very prone to metastasis. The transform growth factor β (TGF-β)-induced epithelial-mesenchymal transition (EMT) is crucially involved in the growth and metastasis of TNBC. This study reported that a natural compound isotoosendanin (ITSN) reduced TNBC metastasis by inhibiting TGF-β-induced EMT and the formation of invadopodia. ITSN can directly interact with TGF-β receptor type-1 (TGFβR1) and abrogated the kinase activity of TGFβR1, thereby blocking the TGF-β-initiated downstream signaling pathway. Moreover, the ITSN-provided inhibition on metastasis obviously disappeared in TGFβR1-overexpressed TNBC cells in vitro as well as in mice bearing TNBC cells overexpressed TGFβR1. Furthermore, Lys232 and Asp351 residues in the kinase domain of TGFβR1 were found to be crucial for the interaction of ITSN with TGFβR1. Additionally, ITSN also improved the inhibitory efficacy of programmed cell death 1 ligand 1 (PD-L1) antibody for TNBC in vivo via inhibiting the TGF-β-mediated EMT in the tumor microenvironment. Our findings not only highlight the key role of TGFβR1 in TNBC metastasis, but also provide a leading compound targeting TGFβR1 for the treatment of TNBC metastasis. Moreover, this study also points out a potential strategy for TNBC treatment by using the combined application of anti-PD-L1 with a TGFβR1 inhibitor.
Collapse
Affiliation(s)
- Jingnan Zhang
- The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines and the SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Ze Zhang
- The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines and the SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Zhenlin Huang
- The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines and the SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Manlin Li
- The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines and the SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Fan Yang
- The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines and the SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Zeqi Wu
- The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines and the SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Qian Guo
- The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines and the SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Xiyu Mei
- The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines and the SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Bin Lu
- The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines and the SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Changhong Wang
- The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines and the SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Zhengtao Wang
- The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines and the SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Lili Ji
- The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines and the SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| |
Collapse
|
38
|
Pandey P, Khan F, Upadhyay TK, Seungjoon M, Park MN, Kim B. New insights about the PDGF/PDGFR signaling pathway as a promising target to develop cancer therapeutic strategies. Biomed Pharmacother 2023; 161:114491. [PMID: 37002577 DOI: 10.1016/j.biopha.2023.114491] [Citation(s) in RCA: 50] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 02/20/2023] [Accepted: 03/07/2023] [Indexed: 03/14/2023] Open
Abstract
Numerous cancers express platelet-derived growth factors (PDGFs) and PDGF receptors (PDGFRs). By directly stimulating tumour cells in an autocrine manner or by stimulating tumour stromal cells in a paracrine manner, the platelet-derived growth factor (PDGF)/platelet-derived growth factor receptor (PDGFR) pathway is crucial in the growth and spread of several cancers. To combat hypoxia in the tumour microenvironment, it encourages angiogenesis. A growing body of experimental data shows that PDGFs target malignant cells, vascular cells, and stromal cells to modulate tumour growth, metastasis, and the tumour microenvironment. To combat medication resistance and enhance patient outcomes in cancers, targeting the PDGF/PDGFR pathway is a viable therapeutic approach. There have been reports of anomalies in the PDGF pathway, including the gain of function point mutations, activating chromosomal translocations, or overexpression or amplification of PDGF receptors (PDGFRs). As a result, it has been shown that targeting the PDGF/PDGFR signaling pathway is an effective method for treating cancer. As a result, this study will concentrate on the regulation of the PDGF/PDGFR signaling system, in particular the current methods and inhibitors used in cancer treatment, as well as the associated therapeutic advantages and side effects.
Collapse
Affiliation(s)
- Pratibha Pandey
- Department of Biotechnology, Noida Institute of Engineering and Technology, Greater Noida, UP, India
| | - Fahad Khan
- Department of Biotechnology, Noida Institute of Engineering and Technology, Greater Noida, UP, India.
| | - Tarun Kumar Upadhyay
- Department of Biotechnology, Parul Institute of Applied Sciences and Centre of Research for Development, Parul University, Vadodara 391760, India
| | - Moon Seungjoon
- Chansol Hospital of Korean Medicine, 290, Buheung-ro, Bupyeong-gu, Incheon 21390, Republic of Korea; Department of Pathology, College of Korean Medicine, Kyung Hee University, Hoegidong Dongdaemun-gu, Seoul 02447, Republic of Korea
| | - Moon Nyeo Park
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Hoegidong Dongdaemun-gu, Seoul 02447, Republic of Korea; Korean Medicine-Based Drug Repositioning Cancer Research Center, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Bonglee Kim
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Hoegidong Dongdaemun-gu, Seoul 02447, Republic of Korea; Korean Medicine-Based Drug Repositioning Cancer Research Center, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea.
| |
Collapse
|
39
|
Hatami M, Kouchak M, Kheirollah A, Khorsandi L, Rashidi M. Effective inhibition of breast cancer stem cell properties by quercetin-loaded solid lipid nanoparticles via reduction of Smad2/Smad3 phosphorylation and β-catenin signaling pathway in triple-negative breast cancer. Biochem Biophys Res Commun 2023; 664:69-76. [PMID: 37141640 DOI: 10.1016/j.bbrc.2023.03.077] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Revised: 03/25/2023] [Accepted: 03/30/2023] [Indexed: 05/06/2023]
Abstract
BACKGROUND The presence of cancer stem cells (CSCs) is a major cause of resistance to cancer therapy and recurrence. Triple-negative breast cancer (TNBC) is a subtype that responds poorly to therapy, making it a significant global health issue. Quercetin (QC) has been shown to affect CSC viability, but its low bioavailability limits its clinical use. This study aims to increase the effectiveness of QC in inhibiting CSC generation by using solid lipid nanoparticles (SLNs) in MDA-MB231 cells. MATERIALS AND METHODS After treating MCF-7 and MDA-MB231 cells with 18.9 μM and 13.4 μM of QC and QC-SLN for 48 h, respectively, cell viability, migration, sphere formation, protein expression of β-catenin, p-Smad 2 and 3, and gene expression of EMT and CSC markers were evaluated. RESULTS The QC-SLN with particle size of 154 nm, zeta potential of -27.7 mV, and encapsulation efficacy of 99.6% was found to be the most effective. Compared to QC, QC-SLN significantly reduced cell viability, migration, sphere formation, protein expression of β-catenin and p-Smad 2 and 3, and gene expression of CD44, zinc finger E-box binding homeobox 1 (ZEB1), vimentin, while increasing the gene expression of E-cadherin. CONCLUSIONS Our findings demonstrate that SLNs improve the cytotoxic effect of QC in MDA-MB231 cells by increasing its bioavailability and inhibiting epithelial-mesenchymal transition (EMT), thereby effectively inhibiting CSC generation. Therefore, SLNs could be a promising new treatment for TNBC, but more in vivo studies are needed to confirm their efficacy.
Collapse
Affiliation(s)
- Mahdi Hatami
- Cellular and Molecular Research Center, Medical Basic Sciences Research Institution, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran; Department of Clinical Biochemistry, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Maryam Kouchak
- Nanotechnology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran; Department of Pharmaceutics, Faculty of Pharmacy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Alireza Kheirollah
- Department of Clinical Biochemistry, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Layasadat Khorsandi
- Cellular and Molecular Research Center, Medical Basic Sciences Research Institution, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mojtaba Rashidi
- Cellular and Molecular Research Center, Medical Basic Sciences Research Institution, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran; Department of Clinical Biochemistry, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| |
Collapse
|
40
|
Guan T, Li M, Song Y, Chen J, Tang J, Zhang C, Wen Y, Yang X, Huang L, Zhu Y, Wang H, Ding K, Zheng J, Zhang H, Liu T. Phosphorylation of USP29 by CDK1 Governs TWIST1 Stability and Oncogenic Functions. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2205873. [PMID: 36782089 PMCID: PMC10104637 DOI: 10.1002/advs.202205873] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Revised: 11/30/2022] [Indexed: 06/18/2023]
Abstract
Triple-negative breast cancer (TNBC) is a highly lethal malignancy with limited therapy options. TWIST1, a key transcriptional factor of epithelial-mesenchymal transition (EMT), contributes to self-renewal of cancer stem-like cells (CSCs), chemo-resistance, metastasis, and TNBC-related death. However, the mechanism by which TWIST1 is deregulated in TNBC remains elusive. Here, USP29 is identified as a bona fide deubiquitinase of TWIST1. The deubiquitination of TWIST1 catalyzed by USP29 is required for its stabilization and subsequent EMT and CSC functions in TNBC, thereby conferring chemotherapeutic resistance and metastasis. Furthermore, the results unexpectedly reveal that CDK1 functions as the direct USP29 activator. Mechanistically, CDK1-mediated phosphorylation of USP29 is essential for its deubiquitinase activity toward TWIST1 and TWIST1 driven-malignant phenotypes in TNBC, which could be markedly mitigated by the genetic ablation or pharmacological inhibition of CDK1. Moreover, the histological analyses show that CDK1 and USP29 are highly upregulated in TNBC samples, which positively correlate with the expression of TWIST1. Taken together, the findings reveal a previously unrecognized tumor-promoting function and clinical significance of the CDK1-USP29 axis through stabilizing TWIST1 and provide the preclinical evidence that targeting this axis is an appealing therapeutic strategy to conquer chemo-resistance and metastasis in TNBC.
Collapse
Affiliation(s)
- Tangming Guan
- College of Pharmacy/International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Ministry of Education (MOE) of ChinaJinan UniversityGuangzhou510632China
| | - Mei Li
- College of Pharmacy/International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Ministry of Education (MOE) of ChinaJinan UniversityGuangzhou510632China
| | - Yan Song
- Department of PathologyNational Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing100021China
| | - Jiayi Chen
- College of Pharmacy/International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Ministry of Education (MOE) of ChinaJinan UniversityGuangzhou510632China
| | - Jiaxin Tang
- Guangdong Provincial Key Laboratory of Genome Stability and Disease PreventionCollege of Life Sciences and OceanographyShenzhen UniversityShenzhen518055China
| | - Caishi Zhang
- College of Pharmacy/International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Ministry of Education (MOE) of ChinaJinan UniversityGuangzhou510632China
| | - Yalei Wen
- College of Pharmacy/International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Ministry of Education (MOE) of ChinaJinan UniversityGuangzhou510632China
| | - Xiao Yang
- College of Pharmacy/International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Ministry of Education (MOE) of ChinaJinan UniversityGuangzhou510632China
| | - Lei Huang
- College of Pharmacy/International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Ministry of Education (MOE) of ChinaJinan UniversityGuangzhou510632China
| | - Yingjie Zhu
- College of Pharmacy/International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Ministry of Education (MOE) of ChinaJinan UniversityGuangzhou510632China
| | - Hongxian Wang
- Department of Thyroid and Breast SurgeryShenzhen Nanshan People's Hospital & The 6th Affiliated Hospital of Shenzhen UniversityShenzhen518052China
| | - Ke Ding
- College of Pharmacy/International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Ministry of Education (MOE) of ChinaJinan UniversityGuangzhou510632China
- State Key Laboratory of Bioorganic and Nature Product ChemistryShanghai Institute of organic chemistryShanghai200032China
| | - Junxia Zheng
- School of Biomedical and Pharmaceutical SciencesGuangdong University of TechnologyGuangzhou510006China
| | - Haoxing Zhang
- Guangdong Provincial Key Laboratory of Genome Stability and Disease PreventionCollege of Life Sciences and OceanographyShenzhen UniversityShenzhen518055China
| | - Tongzheng Liu
- College of Pharmacy/International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Ministry of Education (MOE) of ChinaJinan UniversityGuangzhou510632China
| |
Collapse
|
41
|
Vesuna F, Penet MF, Mori N, Bhujwalla ZM, Raman V. Twist alters the breast tumor microenvironment via choline kinase to facilitate an aggressive phenotype. Mol Cell Biochem 2023; 478:939-948. [PMID: 36136285 PMCID: PMC11299248 DOI: 10.1007/s11010-022-04555-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Accepted: 08/30/2022] [Indexed: 11/26/2022]
Abstract
Twist (TWIST1) is a gene required for cell fate specification in embryos and its expression in mammary epithelium can initiate tumorigenesis through the epithelial-mesenchymal transition. To identify downstream target genes of Twist in breast cancer, we performed microarray analysis on the transgenic breast cancer cell line, MCF-7/Twist. One of the targets identified was choline kinase whose upregulation resulted in increased cellular phosphocholine and total choline containing compounds-a characteristic observed in highly aggressive metastatic cancers. To study the interactions between Twist, choline kinase, and their effect on the microenvironment, we used 1H magnetic resonance spectroscopy and found significantly higher phosphocholine and total choline, as well as increased phosphocholine/glycerophosphocholine ratio in MCF-7/Twist cells. We also observed significant increases in extracellular glucose, lactate, and [H +] ion concentrations in the MCF-7/Twist cells. Magnetic resonance imaging of MCF-7/Twist orthotopic breast tumors showed a significant increase in vascular volume and permeability surface area product compared to control tumors. In addition, by reverse transcription-quantitative polymerase chain reaction, we discovered that Twist upregulated choline kinase expression in estrogen receptor negative breast cancer cell lines through FOXA1 downregulation. Moreover, using The Cancer Genome Atlas database, we observed a significant inverse relationship between FOXA1 and choline kinase expression and propose that it could act as a modulator of the Twist/choline kinase axis. The data presented indicate that Twist is a driver of choline kinase expression in breast cancer cells via FOXA1 resulting in the generation of an aggressive breast cancer phenotype.
Collapse
Affiliation(s)
- Farhad Vesuna
- Division of Cancer Imaging Research, Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Marie-France Penet
- Division of Cancer Imaging Research, Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Noriko Mori
- Division of Cancer Imaging Research, Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Zaver M Bhujwalla
- Division of Cancer Imaging Research, Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Venu Raman
- Division of Cancer Imaging Research, Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands.
- Department of Pharmacology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
42
|
Zhou T, Zhang LY, He JZ, Miao ZM, Li YY, Zhang YM, Liu ZW, Zhang SZ, Chen Y, Zhou GC, Liu YQ. Review: Mechanisms and perspective treatment of radioresistance in non-small cell lung cancer. Front Immunol 2023; 14:1133899. [PMID: 36865554 PMCID: PMC9971010 DOI: 10.3389/fimmu.2023.1133899] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Accepted: 01/31/2023] [Indexed: 02/16/2023] Open
Abstract
Radiotherapy is the major treatment of non-small cell lung cancer (NSCLC). The radioresistance and toxicity are the main obstacles that leading to therapeutic failure and poor prognosis. Oncogenic mutation, cancer stem cells (CSCs), tumor hypoxia, DNA damage repair, epithelial-mesenchymal transition (EMT), and tumor microenvironment (TME) may dominate the occurrence of radioresistance at different stages of radiotherapy. Chemotherapy drugs, targeted drugs, and immune checkpoint inhibitors are combined with radiotherapy to treat NSCLC to improve the efficacy. This article reviews the potential mechanism of radioresistance in NSCLC, and discusses the current drug research to overcome radioresistance and the advantages of Traditional Chinese medicine (TCM) in improving the efficacy and reducing the toxicity of radiotherapy.
Collapse
Affiliation(s)
- Ting Zhou
- Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and The Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and University, Gansu University of Chinese Medicine, Lanzhou, China,Experimental & Training Teaching Centers, Gansu University of Chinese Medicine, Lanzhou, China
| | - Li-Ying Zhang
- Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and The Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and University, Gansu University of Chinese Medicine, Lanzhou, China,College of Basic Medicine, Gansu University of Chinese Medicine, Lanzhou, China
| | - Jian-Zheng He
- Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and The Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and University, Gansu University of Chinese Medicine, Lanzhou, China,College of Basic Medicine, Gansu University of Chinese Medicine, Lanzhou, China
| | - Zhi-Ming Miao
- Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and The Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and University, Gansu University of Chinese Medicine, Lanzhou, China
| | - Yang-Yang Li
- Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and The Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and University, Gansu University of Chinese Medicine, Lanzhou, China
| | - Yi-Ming Zhang
- Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and The Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and University, Gansu University of Chinese Medicine, Lanzhou, China
| | - Zhi-Wei Liu
- Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and The Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and University, Gansu University of Chinese Medicine, Lanzhou, China
| | - Shang-Zu Zhang
- Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and The Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and University, Gansu University of Chinese Medicine, Lanzhou, China
| | - Yan Chen
- Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and The Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and University, Gansu University of Chinese Medicine, Lanzhou, China
| | - Gu-Cheng Zhou
- Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and The Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and University, Gansu University of Chinese Medicine, Lanzhou, China
| | - Yong-Qi Liu
- Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and The Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and University, Gansu University of Chinese Medicine, Lanzhou, China,College of Basic Medicine, Gansu University of Chinese Medicine, Lanzhou, China,Key Laboratory of Dunhuang Medicine and Transformation at Provincial and Ministerial Level, Gansu University of Chinese Medicine, Lanzhou, China,*Correspondence: Yong-Qi Liu,
| |
Collapse
|
43
|
Mathiesen A, Haynes B, Huyck R, Brown M, Dobrian A. Adipose Tissue-Derived Extracellular Vesicles Contribute to Phenotypic Plasticity of Prostate Cancer Cells. Int J Mol Sci 2023; 24:1229. [PMID: 36674745 PMCID: PMC9864182 DOI: 10.3390/ijms24021229] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 01/03/2023] [Accepted: 01/05/2023] [Indexed: 01/11/2023] Open
Abstract
Metastatic prostate cancer is one of the leading causes of male cancer deaths in the western world. Obesity significantly increases the risk of metastatic disease and is associated with a higher mortality rate. Systemic chronic inflammation can result from a variety of conditions, including obesity, where adipose tissue inflammation is a major contributor. Adipose tissue endothelial cells (EC) exposed to inflammation become dysfunctional and produce a secretome, including extracellular vesicles (EV), that can impact function of cells in distant tissues, including malignant cells. The aim of this study was to explore the potential role of EVs produced by obese adipose tissue and the ECs exposed to pro-inflammatory cytokines on prostate cancer phenotypic plasticity in vitro. We demonstrate that PC3ML metastatic prostate cancer cells exposed to EVs from adipose tissue ECs and to EVs from human adipose tissue total explants display reduced invasion and increased proliferation. The latter functional changes could be attributed to the EV miRNA cargo. We also show that the functional shift is TWIST1-dependent and is consistent with mesenchymal-to-epithelial transition, which is key to establishment of secondary tumor growth. Understanding the complex effects of EVs on prostate cancer cells of different phenotypes is key before their intended use as therapeutics.
Collapse
Affiliation(s)
- Allison Mathiesen
- Department of Physiological Sciences, Eastern Virginia Medical School, Norfolk, VA 23501, USA
| | - Bronson Haynes
- Department of Physiological Sciences, Eastern Virginia Medical School, Norfolk, VA 23501, USA
| | - Ryan Huyck
- Department of Physiological Sciences, Eastern Virginia Medical School, Norfolk, VA 23501, USA
| | - Michael Brown
- Department of Physiological Sciences, Eastern Virginia Medical School, Norfolk, VA 23501, USA
| | - Anca Dobrian
- Department of Physiological Sciences, Eastern Virginia Medical School, Norfolk, VA 23501, USA
| |
Collapse
|
44
|
Zhang J, Hu Z, Horta CA, Yang J. Regulation of epithelial-mesenchymal transition by tumor microenvironmental signals and its implication in cancer therapeutics. Semin Cancer Biol 2023; 88:46-66. [PMID: 36521737 DOI: 10.1016/j.semcancer.2022.12.002] [Citation(s) in RCA: 45] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 12/06/2022] [Accepted: 12/08/2022] [Indexed: 12/15/2022]
Abstract
Epithelial-mesenchymal transition (EMT) has been implicated in various aspects of tumor development, including tumor invasion and metastasis, cancer stemness, and therapy resistance. Diverse stroma cell types along with biochemical and biophysical factors in the tumor microenvironment impinge on the EMT program to impact tumor progression. Here we provide an in-depth review of various tumor microenvironmental signals that regulate EMT in cancer. We discuss the molecular mechanisms underlying the role of EMT in therapy resistance and highlight new therapeutic approaches targeting the tumor microenvironment to impact EMT and tumor progression.
Collapse
Affiliation(s)
- Jing Zhang
- Department of Pharmacology, Moores Cancer Center, University of California, San Diego, School of Medicine, La Jolla, CA 92093, USA
| | - Zhimin Hu
- Department of Pharmacology, Moores Cancer Center, University of California, San Diego, School of Medicine, La Jolla, CA 92093, USA
| | - Calista A Horta
- Department of Pharmacology, Moores Cancer Center, University of California, San Diego, School of Medicine, La Jolla, CA 92093, USA
| | - Jing Yang
- Department of Pharmacology, Moores Cancer Center, University of California, San Diego, School of Medicine, La Jolla, CA 92093, USA; Department of Pediatrics, University of California, San Diego, School of Medicine, La Jolla, CA 92093, USA.
| |
Collapse
|
45
|
Twafra S, Sokolik CG, Sneh T, Srikanth KD, Meirson T, Genna A, Chill JH, Gil-Henn H. A novel Pyk2-derived peptide inhibits invadopodia-mediated breast cancer metastasis. Oncogene 2023; 42:278-292. [PMID: 36258022 DOI: 10.1038/s41388-022-02481-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2022] [Revised: 09/03/2022] [Accepted: 09/21/2022] [Indexed: 01/28/2023]
Abstract
Dissemination of cancer cells from the primary tumor into distant body tissues and organs is the leading cause of death in cancer patients. While most clinical strategies aim to reduce or impede the growth of the primary tumor, no treatment to eradicate metastatic cancer exists at present. Metastasis is mediated by feet-like cytoskeletal structures called invadopodia which allow cells to penetrate through the basement membrane and intravasate into blood vessels during their spread to distant tissues and organs. The non-receptor tyrosine kinase Pyk2 is highly expressed in breast cancer, where it mediates invadopodia formation and function via interaction with the actin-nucleation-promoting factor cortactin. Here, we designed a cell-permeable peptide inhibitor that contains the second proline-rich region (PRR2) sequence of Pyk2, which binds to the SH3 domain of cortactin and inhibits the interaction between Pyk2 and cortactin in invadopodia. The Pyk2-PRR2 peptide blocks spontaneous lung metastasis in immune-competent mice by inhibiting cortactin tyrosine phosphorylation and actin polymerization-mediated maturation and activation of invadopodia, leading to reduced MMP-dependent tumor cell invasiveness. The native structure of the Pyk2-PRR2:cortactin-SH3 complex was determined using nuclear magnetic resonance (NMR), revealing an extended class II interaction surface spanning the canonical binding groove and a second hydrophobic surface which significantly contributes to ligand affinity. Using structure-guided design, we created a mutant peptide lacking critical residues involved in binding that failed to inhibit invadopodia maturation and function and consequent metastatic dissemination in mice. Our findings shed light on the specific molecular interactions between Pyk2 and cortactin and may lead to the development of novel strategies for preventing dissemination of primary breast tumors predicted at the time of diagnosis to be highly metastatic, and of secondary tumors that have already spread to other parts of the body.
Collapse
Affiliation(s)
- Shams Twafra
- Cell Migration and Invasion Laboratory, The Azrieli Faculty of Medicine, Bar-Ilan University, Safed, 1311502, Israel
| | - Chana G Sokolik
- Bio-NMR Laboratory, Department of Chemistry, Bar-Ilan University, Ramat-Gan, 52900, Israel
| | - Tal Sneh
- Cell Migration and Invasion Laboratory, The Azrieli Faculty of Medicine, Bar-Ilan University, Safed, 1311502, Israel
| | - Kolluru D Srikanth
- Cell Migration and Invasion Laboratory, The Azrieli Faculty of Medicine, Bar-Ilan University, Safed, 1311502, Israel
| | - Tomer Meirson
- Cell Migration and Invasion Laboratory, The Azrieli Faculty of Medicine, Bar-Ilan University, Safed, 1311502, Israel.,Davidoff Cancer Center, Rabin Medical Center-Beilinson Hospital, Petah Tikva, Israel
| | - Alessandro Genna
- Cell Migration and Invasion Laboratory, The Azrieli Faculty of Medicine, Bar-Ilan University, Safed, 1311502, Israel
| | - Jordan H Chill
- Bio-NMR Laboratory, Department of Chemistry, Bar-Ilan University, Ramat-Gan, 52900, Israel.
| | - Hava Gil-Henn
- Cell Migration and Invasion Laboratory, The Azrieli Faculty of Medicine, Bar-Ilan University, Safed, 1311502, Israel.
| |
Collapse
|
46
|
Perrin L, Belova E, Bayarmagnai B, Tüzel E, Gligorijevic B. Invadopodia enable cooperative invasion and metastasis of breast cancer cells. Commun Biol 2022; 5:758. [PMID: 35915226 PMCID: PMC9343607 DOI: 10.1038/s42003-022-03642-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Accepted: 06/28/2022] [Indexed: 11/29/2022] Open
Abstract
Invasive and non-invasive cancer cells can invade together during cooperative invasion. However, the events leading to it, role of the epithelial-mesenchymal transition and the consequences this may have on metastasis are unknown. In this study, we demonstrate that the isogenic 4T1 and 67NR breast cancer cells sort from each other in 3D spheroids, followed by cooperative invasion. By time-lapse microscopy, we show that the invasive 4T1 cells move more persistently compared to non-invasive 67NR, sorting and accumulating at the spheroid-matrix interface, a process dependent on cell-matrix adhesions and independent from E-cadherin cell-cell adhesions. Elimination of invadopodia in 4T1 cells blocks invasion, demonstrating that invadopodia requirement is limited to leader cells. Importantly, we demonstrate that cells with and without invadopodia can also engage in cooperative metastasis in preclinical mouse models. Altogether, our results suggest that a small number of cells with invadopodia can drive the metastasis of heterogeneous cell clusters. Cooperative invasion requires the formation of invadopodia in the leader cells, and a small number of leader cells may be enough to facilitate cooperative invasion and metastasis, including non-invadopodia forming cancer cells.
Collapse
|
47
|
Sun X, Zhou L, Wang X, Li Y, Liu X, Chen Y, Zhong Z, Chen J. FYCO1 regulates migration, invasion, and invadopodia formation in HeLa cells through CDC42/N-WASP/Arp2/3 signaling pathway. Biochem Cell Biol 2022; 100:458-472. [PMID: 36342046 DOI: 10.1139/bcb-2021-0575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
FYCO1, an autophagy adaptor, plays an essential role in the trafficking toward the plus-end of microtubules and the fusion of autophagosomes. Autophagic dysfunction is involved in numerous disease states, including cancers. Previous studies have implicated FYCO1 as one of the critical genes involved in the adenoma to carcinoma transition, but the biological function and mechanism of FYCO1 in carcinogenesis remain unclear. This study aims to elucidate the role and mechanism of up- and downregulation of FYCO1 in mediating tumor effects in HeLa cells. Functionally, FYCO1 promotes cellular migration, invasion, epithelial-mesenchymal transition, invadopodia formation, and matrix degradation, which are detected through wound healing, transwell, immunofluorescence, and Western blot approaches. Interestingly, the data show that although FYCO1 does not affect HeLa cell proliferation, cell cycle distribution, nor vessels' formation, FYCO1 can block the apoptotic function. FYCO1 inhibits cleavage of PARP, caspase3, and caspase9 and increases Bcl-2/Bax ratio. Then, we used CK666, an Arp2/3 specific inhibitor, to confirm that FYCO1 may promote the migration and invasion of HeLa cells through the CDC42/N-WASP/Arp2/3 signaling pathway. Taken together, these results provide a new insight that FYCO1, an autophagy adaptor, may also be a new regulator of tumor metastasis.
Collapse
Affiliation(s)
- Xuejiao Sun
- Translational Research Institute of Brain and Brain-like Intelligence, People's Hospital, School of Medicine, Tongji University, Shanghai 200434, China.,Department of Pediatrics, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai 200434, China.,Department of Medical Genetics, School of Medicine, Tongji University, Shanghai 200092, China.,Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai 200434, China
| | - Linlin Zhou
- Translational Research Institute of Brain and Brain-like Intelligence, People's Hospital, School of Medicine, Tongji University, Shanghai 200434, China.,Department of Pediatrics, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai 200434, China.,Department of Medical Genetics, School of Medicine, Tongji University, Shanghai 200092, China.,Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai 200434, China
| | - Xinyao Wang
- Translational Research Institute of Brain and Brain-like Intelligence, People's Hospital, School of Medicine, Tongji University, Shanghai 200434, China.,Department of Pediatrics, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai 200434, China.,Department of Medical Genetics, School of Medicine, Tongji University, Shanghai 200092, China.,Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai 200434, China
| | - Yuying Li
- Translational Research Institute of Brain and Brain-like Intelligence, People's Hospital, School of Medicine, Tongji University, Shanghai 200434, China.,Department of Pediatrics, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai 200434, China.,Department of Medical Genetics, School of Medicine, Tongji University, Shanghai 200092, China.,Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai 200434, China
| | - Xiangyuan Liu
- Translational Research Institute of Brain and Brain-like Intelligence, People's Hospital, School of Medicine, Tongji University, Shanghai 200434, China.,Department of Pediatrics, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai 200434, China.,Department of Medical Genetics, School of Medicine, Tongji University, Shanghai 200092, China.,Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai 200434, China
| | - Yu Chen
- Department of Pediatrics, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai 200434, China
| | - Zilin Zhong
- Translational Research Institute of Brain and Brain-like Intelligence, People's Hospital, School of Medicine, Tongji University, Shanghai 200434, China.,Department of Pediatrics, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai 200434, China.,Department of Medical Genetics, School of Medicine, Tongji University, Shanghai 200092, China.,Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai 200434, China
| | - Jianjun Chen
- Translational Research Institute of Brain and Brain-like Intelligence, People's Hospital, School of Medicine, Tongji University, Shanghai 200434, China.,Department of Pediatrics, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai 200434, China.,Department of Medical Genetics, School of Medicine, Tongji University, Shanghai 200092, China.,Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai 200434, China
| |
Collapse
|
48
|
Sun Y, Qu H, Song Q, Shen Y, Wang L, Niu X. High-glucose induced toxicity in HK-2 cells can be alleviated by inhibition of miRNA-320c. Ren Fail 2022; 44:1388-1398. [PMID: 35969018 PMCID: PMC9389931 DOI: 10.1080/0886022x.2022.2106874] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Diabetic nephropathy (DN) is a major healthcare challenge worldwide. MiRNAs exert a regulatory effect on the progress of DN. Our study proposed to investigate the miR-320c expression and its function on the pathogenesis of DN in vitro. The level of miR-320c in HK-2 cells was quantified by RT-qPCR. Cell morphology, invasion, and migration were observed by optical microscope, Transwell invasion assay, and scratch wound assay. Then, the levels of PTEN, α-SMA, vimentin, E-cadherin, p-PI3K, PI3K, AKT, and p-AKT were analyzed through western blotting. A Dual-luciferase reporter assay was conducted to explore the target relationship between miR-320c and PTEN. It was discovered that miR-320c was over-expressed in high glucose (HG)-treated HK-2 cells. Furthermore, inhibition of miR-320c could alleviate the epithelial-mesenchymal transition (EMT) of HG-induced HK-2 cells and retain the normal morphology of HK-2 cells. Additionally, the miR-320c inhibitor decreased the invasiveness and migration of HG-treated HK-2 cells. Next, the target gene of miR-320c, PTEN, was identified, and the function of miR-320c was reversed by down-regulation of PTEN. Finally, we found inhibition of miR-320c restrained the PI3K/AKT pathway. Therefore, inhibition of miR-320c could alleviate toxicity of HK-2 cells induced by HG via targeting PTEN and restraining the PI3K/AKT pathway, illustrating that miR-320c may act as a new biomarker in the diagnosis of DN.
Collapse
Affiliation(s)
- Yan Sun
- Heji Hospital Affiliated to Changzhi Medical College, Changzhi, China
| | - Hai Qu
- Department of General Surgery, Heji Hospital Affiliated to Changzhi Medical College, Changzhi, China
| | - Qi Song
- Heji Hospital Affiliated to Changzhi Medical College, Changzhi, China
| | - Yifan Shen
- Heji Hospital Affiliated to Changzhi Medical College, Changzhi, China
| | - Lijuan Wang
- Heji Hospital Affiliated to Changzhi Medical College, Changzhi, China
| | - Xiaohong Niu
- Heji Hospital Affiliated to Changzhi Medical College, Changzhi, China
| |
Collapse
|
49
|
Chen B, Wang M, Qiu J, Liao K, Zhang W, Lv Q, Ma C, Qian Z, Shi Z, Liang R, Lin Y, Ye J, Qiu Y, Lin Y. Cleavage of tropomodulin-3 by asparagine endopeptidase promotes cancer malignancy by actin remodeling and SND1/RhoA signaling. J Exp Clin Cancer Res 2022; 41:209. [PMID: 35765111 PMCID: PMC9238189 DOI: 10.1186/s13046-022-02411-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Accepted: 06/07/2022] [Indexed: 12/12/2022] Open
Abstract
Abstract
Background
Abnormal proliferation and migration of cells are hallmarks of cancer initiation and malignancy. Asparagine endopeptidase (AEP) has specific substrate cleavage ability and plays a pro-cancer role in a variety of cancers. However, the underlying mechanism of AEP in cancer proliferation and migration still remains unclear.
Methods
Co-immunoprecipitation and following mass spectrometry were used to identify the substrate of AEP. Western blotting was applied to measure the expression of proteins. Single cell/nuclear-sequences were done to detect the heterogeneous expression of Tmod3 in tumor tissues. CCK-8 assay, flow cytometry assays, colony formation assay, Transwell assay and scratch wound-healing assay were performed as cellular functional experiments. Mouse intracranial xenograft tumors were studied in in vivo experiments.
Results
Here we showed that AEP cleaved a ubiquitous cytoskeleton regulatory protein, tropomodulin-3 (Tmod3) at asparagine 157 (N157) and produced two functional truncations (tTmod3-N and tTmod3-C). Truncated Tmod3 was detected in diverse tumors and was found to be associated with poor prognosis of high-grade glioma. Functional studies showed that tTmod3-N and tTmod3-C enhanced cancer cell migration and proliferation, respectively. Animal models further revealed the tumor-promoting effects of AEP truncated Tmod3 in vivo. Mechanistically, tTmod3-N was enriched in the cell cortex and competitively inhibited the pointed-end capping effect of wild-type Tmod3 on filamentous actin (F-actin), leading to actin remodeling. tTmod3-C translocated to the nucleus, where it interacted with Staphylococcal Nuclease And Tudor Domain Containing 1 (SND1), facilitating the transcription of Ras Homolog Family Member A/Cyclin Dependent Kinases (RhoA/CDKs).
Conclusion
The newly identified AEP-Tmod3 protease signaling axis is a novel “dual-regulation” mechanism of tumor cell proliferation and migration. Our work provides new clues to the underlying mechanisms of cancer proliferation and invasive progression and evidence for targeting AEP or Tmod3 for therapy.
Collapse
|
50
|
Perrin L, Gligorijevic B. Proteolytic and mechanical remodeling of the extracellular matrix by invadopodia in cancer. Phys Biol 2022; 20:10.1088/1478-3975/aca0d8. [PMID: 36343366 PMCID: PMC9942491 DOI: 10.1088/1478-3975/aca0d8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 11/07/2022] [Indexed: 11/09/2022]
Abstract
Cancer invasion and metastasis require remodeling of the adjacent extracellular matrix (ECM). In this mini review, we will cover the mechanisms of proteolytic degradation and the mechanical remodeling of the ECM by cancer cells, with a focus on invadopodia. Invadopodia are membrane protrusions unique to cancer cells, characterized by an actin core and by the focal degradation of ECM via matrix metalloproteases (MMPs). While ECM can also be remodeled, at lower levels, by focal adhesions, or internal collagen digestion, invadopodia are now recognized as the major mechanism for MMP-dependent pericellular ECM degradation by cancer cells. Recent evidence suggests that the completion of epithelial-mesenchymal transition may be dispensable for invadopodia and metastasis, and that invadopodia are required not only for mesenchymal, single cell invasion, but also for collective invasion. During collective invasion, invadopodia was then shown to be located in leader cells, allowing follower cells to move via cooperation. Collectively, this suggests that invadopodia function may be a requirement not only for later steps of metastasis, but also for early invasion of epithelial cells into the stromal tissue. Over the last decade, invadopodia studies have transitioned into in 3D andin vivosettings, leading to the confirmation of their essential role in metastasis in preclinical animal models. In summary, invadopodia may hold a great potential for individual risk assessment as a prognostic marker for metastasis, as well as a therapeutic target.
Collapse
Affiliation(s)
- L. Perrin
- Bioengineering Department, Temple University, Philadelphia PA, USA
- Present address, Institut Curie, Paris, France
| | - B. Gligorijevic
- Bioengineering Department, Temple University, Philadelphia PA, USA
- Cancer Signaling and Epigenetics Program, Fox Chase Cancer Center, Philadelphia PA, USA
| |
Collapse
|