1
|
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
|
2
|
Sankhe CS, Sacco JL, Lawton J, Fair RA, Soares DVR, Aldahdooh MKR, Gomez ED, Gomez EW. Breast Cancer Cells Exhibit Mesenchymal-Epithelial Plasticity Following Dynamic Modulation of Matrix Stiffness. Adv Biol (Weinh) 2024; 8:e2400087. [PMID: 38977422 DOI: 10.1002/adbi.202400087] [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: 02/17/2024] [Revised: 05/31/2024] [Indexed: 07/10/2024]
Abstract
Mesenchymal-epithelial transition (MET) is essential for tissue and organ development and is thought to contribute to cancer by enabling the establishment of metastatic lesions. Despite its importance in both health and disease, there is a lack of in vitro platforms to study MET and little is known about the regulation of MET by mechanical cues. Here, hyaluronic acid-based hydrogels with dynamic and tunable stiffnesses mimicking that of normal and tumorigenic mammary tissue are synthesized. The platform is then utilized to examine the response of mammary epithelial cells and breast cancer cells to dynamic modulation of matrix stiffness. Gradual softening of the hydrogels reduces proliferation and increases apoptosis of breast cancer cells. Moreover, breast cancer cells exhibit temporal changes in cell morphology, cytoskeletal organization, and gene expression that are consistent with mesenchymal-epithelial plasticity as the stiffness of the matrix is reduced. A reduction in matrix stiffness attenuates the expression of integrin-linked kinase, and inhibition of integrin-linked kinase impacts proliferation, apoptosis, and gene expression in cells cultured on stiff and dynamic hydrogels. Overall, these findings reveal intermediate epithelial/mesenchymal states as cells move along a matrix stiffness-mediated MET trajectory and suggest an important role for matrix mechanics in regulating mesenchymal-epithelial plasticity.
Collapse
Affiliation(s)
- Chinmay S Sankhe
- Department of Chemical Engineering, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Jessica L Sacco
- Department of Chemical Engineering, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Jacob Lawton
- Department of Chemical Engineering, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Ryan A Fair
- Department of Materials Science and Engineering, The Pennsylvania State University, University Park, PA, 16802, USA
| | | | - Mohammed K R Aldahdooh
- Department of Chemistry, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Enrique D Gomez
- Department of Chemical Engineering, The Pennsylvania State University, University Park, PA, 16802, USA
- Department of Materials Science and Engineering, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Esther W Gomez
- Department of Chemical Engineering, The Pennsylvania State University, University Park, PA, 16802, USA
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA, 16802, USA
| |
Collapse
|
3
|
Frederick MI, Abdesselam D, Clouvel A, Croteau L, Hassan S. Leveraging PARP-1/2 to Target Distant Metastasis. Int J Mol Sci 2024; 25:9032. [PMID: 39201718 PMCID: PMC11354653 DOI: 10.3390/ijms25169032] [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: 07/22/2024] [Revised: 08/14/2024] [Accepted: 08/14/2024] [Indexed: 09/03/2024] Open
Abstract
Poly (ADP-Ribose) Polymerase (PARP) inhibitors have changed the outcomes and therapeutic strategy for several cancer types. As a targeted therapeutic mainly for patients with BRCA1/2 mutations, PARP inhibitors have commonly been exploited for their capacity to prevent DNA repair. In this review, we discuss the multifaceted roles of PARP-1 and PARP-2 beyond DNA repair, including the impact of PARP-1 on chemokine signalling, immune modulation, and transcriptional regulation of gene expression, particularly in the contexts of angiogenesis and epithelial-to-mesenchymal transition (EMT). We evaluate the pre-clinical role of PARP inhibitors, either as single-agent or combination therapies, to block the metastatic process. Efficacy of PARP inhibitors was demonstrated via DNA repair-dependent and independent mechanisms, including DNA damage, cell migration, invasion, initial colonization at the metastatic site, osteoclastogenesis, and micrometastasis formation. Finally, we summarize the recent clinical advancements of PARP inhibitors in the prevention and progression of distant metastases, with a particular focus on specific metastatic sites and PARP-1 selective inhibitors. Overall, PARP inhibitors have demonstrated great potential in inhibiting the metastatic process, pointing the way for greater use in early cancer settings.
Collapse
Affiliation(s)
- Mallory I. Frederick
- Faculty of Medicine, Université de Montréal, Montreal, QC H3C 3T5, Canada; (M.I.F.); (D.A.); (L.C.)
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), l’Institut de Cancer de Montreal, Montreal, QC H2X 0A9, Canada;
| | - Djihane Abdesselam
- Faculty of Medicine, Université de Montréal, Montreal, QC H3C 3T5, Canada; (M.I.F.); (D.A.); (L.C.)
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), l’Institut de Cancer de Montreal, Montreal, QC H2X 0A9, Canada;
| | - Anna Clouvel
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), l’Institut de Cancer de Montreal, Montreal, QC H2X 0A9, Canada;
| | - Laurent Croteau
- Faculty of Medicine, Université de Montréal, Montreal, QC H3C 3T5, Canada; (M.I.F.); (D.A.); (L.C.)
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), l’Institut de Cancer de Montreal, Montreal, QC H2X 0A9, Canada;
| | - Saima Hassan
- Faculty of Medicine, Université de Montréal, Montreal, QC H3C 3T5, Canada; (M.I.F.); (D.A.); (L.C.)
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), l’Institut de Cancer de Montreal, Montreal, QC H2X 0A9, Canada;
- Division of Surgical Oncology, Department of Surgery, Centre Hospitalier de l’Université de Montréal (CHUM), Montreal, QC H2X 0C1, Canada
| |
Collapse
|
4
|
Kong D, Zha L, Yao Y, Zhang Z, Gao J, Zhang R, Chen L, Wang Z. Effects of HMGA2 on the biological characteristics and stemness acquisition of gastric cancer cells. Arab J Gastroenterol 2024; 25:135-142. [PMID: 38378354 DOI: 10.1016/j.ajg.2024.01.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Revised: 09/01/2023] [Accepted: 01/02/2024] [Indexed: 02/22/2024]
Abstract
BACKGROUND AND STUDY AIMS The high mobility group A2 (HMGA2), a nonhistone nuclear binding protein, modulates transcription by altering the chromatin architecture of the target gene DNA in its specific AT-hooks region. HMGA2 overexpression has been observed in embryonic tissue and many malignant neoplasms. This study sought to verify whether HMGA2 plays a role in the biological functions of gastric cancer cells, such as cell proliferation, invasiveness, migration, and stem cell acquisition, and to provide some ideas for further research on the metastatic mechanism of gastric cancer. PATIENTS AND METHODS HMGA2's effects on the proliferation, invasiveness, and migration capabilities of gastric cancer cells were individually detected by BrdU, Transwell, and wound healing assays. Western blotting and immunofluorescence were used to evaluate whether HMGA2 could promote the acquisition of gastric cancer cells. Biostatistical analyses were performed using SPSS 17.0 for Windows. RESULTS HMGA2 expression levels in gastric cancer cell lines were significantly higher than those in human immortalized gastric epithelial cell lines (p < 0.01). Gastric cancer cell proliferation was inhibited when HMGA2 was overexpressed (p < 0.05). The invasiveness and migration capabilities of gastric cancer cells with HMGA2 overexpression were enhanced more than those of the corresponding control groups (p < 0.05). HMGA2 overexpression promotes the stemness acquisition of stem cells from gastric cancer cells. CONCLUSIONS This study verified that the HMGA2 structural transcription factor promotes invasiveness, migration, and acquisition of gastric cancer cells. Furthermore, our findings provide significant insight for further research on the metastatic mechanism of gastric cancer.
Collapse
Affiliation(s)
- Dequan Kong
- Department of Emergency Medicine, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361101, China.
| | - Lang Zha
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Yaben Yao
- Department of Emergency Medicine, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361101, China
| | - Zhenyu Zhang
- Department of Emergency Medicine, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361101, China
| | - Jun Gao
- Department of Emergency Medicine, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361101, China
| | - Rui Zhang
- Department of Emergency Medicine, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361101, China
| | - Lei Chen
- Department of Emergency Medicine, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361101, China
| | - Ziwei Wang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| |
Collapse
|
5
|
Jasim SA, Al-Hawary SIS, Hjazi A, Ahmad I, Kaur I, Kadhum WR, Alkhafaji AT, Ghildiyal P, Jawad MA, Alsaadi SB. A comprehensive review of lncRNA CRNDE in cancer progression and pathology, with a specific glance at the epithelial-mesenchymal transition (EMT) process. Pathol Res Pract 2024; 256:155229. [PMID: 38484655 DOI: 10.1016/j.prp.2024.155229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Revised: 02/09/2024] [Accepted: 02/25/2024] [Indexed: 04/14/2024]
Abstract
It has been suggested that the long non-coding RNAs (lncRNAs), such as colorectal neoplasia differentially expressed (CRNDE), may contribute to the formation of human cancer. It is yet unknown, though, what therapeutic significance CRNDE expression has for different forms of cancer. CRNDE has recently been proposed as a possible diagnostic biomarker and prognostic pred for excellent specificity and sensitivity in cancer tissues and plasma. To provide the groundwork for potential future therapeutic uses of CRNDE, we briefly overview its biological action and related cancer-related pathways. Next, we mainly address the impact of CRNDE on the epithelial-mesenchymal transition (EMT). The epithelial-mesenchymal transition, or EMT, is an essential biological mechanism involved in the spread of cancer.
Collapse
Affiliation(s)
| | | | - Ahmed Hjazi
- Department of Medical Laboratory, College of Applied Medical Sciences, Prince Sattam bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia
| | - Irfan Ahmad
- Department of Clinical Laboratory Sciences, College of Applied Medical Science, King Khalid University, Abha, Saudi Arabia.
| | - Irwanjot Kaur
- Department of Biotechnology and Genetics, Jain (Deemed-to-be) University, Bengaluru, Karnataka 560069, India; Department of Allied Healthcare and Sciences, Vivekananda Global University, Jaipur, Rajasthan 303012, India
| | - Wesam R Kadhum
- Department of Pharmacy, Kut University College, Kut, Wasit 52001, Iraq; Advanced research center, Kut University College, Kut, Wasit 52001, Iraq
| | | | - Pallavi Ghildiyal
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | | | - Salim B Alsaadi
- Department of Pharmaceutics, Al-Hadi University College, Baghdad 10011, Iraq
| |
Collapse
|
6
|
Subramanian A, Nemat-Gorgani N, Ellis-Caleo TJ, van IJzendoorn DGP, Sears TJ, Somani A, Luca BA, Zhou MY, Bradic M, Torres IA, Oladipo E, New C, Kenney DE, Avedian RS, Steffner RJ, Binkley MS, Mohler DG, Tap WD, D'Angelo SP, van de Rijn M, Ganjoo KN, Bui NQ, Charville GW, Newman AM, Moding EJ. Sarcoma microenvironment cell states and ecosystems are associated with prognosis and predict response to immunotherapy. NATURE CANCER 2024; 5:642-658. [PMID: 38429415 PMCID: PMC11058033 DOI: 10.1038/s43018-024-00743-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/24/2023] [Accepted: 02/08/2024] [Indexed: 03/03/2024]
Abstract
Characterization of the diverse malignant and stromal cell states that make up soft tissue sarcomas and their correlation with patient outcomes has proven difficult using fixed clinical specimens. Here, we employed EcoTyper, a machine-learning framework, to identify the fundamental cell states and cellular ecosystems that make up sarcomas on a large scale using bulk transcriptomes with clinical annotations. We identified and validated 23 sarcoma-specific, transcriptionally defined cell states, many of which were highly prognostic of patient outcomes across independent datasets. We discovered three conserved cellular communities or ecotypes associated with underlying genomic alterations and distinct clinical outcomes. We show that one ecotype defined by tumor-associated macrophages and epithelial-like malignant cells predicts response to immune-checkpoint inhibition but not chemotherapy and validate our findings in an independent cohort. Our results may enable identification of patients with soft tissue sarcomas who could benefit from immunotherapy and help develop new therapeutic strategies.
Collapse
Affiliation(s)
- Ajay Subramanian
- Department of Radiation Oncology, Stanford University, Stanford, CA, USA
| | - Neda Nemat-Gorgani
- Department of Radiation Oncology, Stanford University, Stanford, CA, USA
| | | | | | - Timothy J Sears
- Department of Radiation Oncology, Stanford University, Stanford, CA, USA
| | - Anish Somani
- Department of Radiation Oncology, Stanford University, Stanford, CA, USA
| | - Bogdan A Luca
- Department of Biomedical Data Science, Stanford University, Stanford, CA, USA
| | - Maggie Y Zhou
- Division of Oncology, Department of Medicine, Stanford University, Stanford, CA, USA
| | - Martina Bradic
- Marie-Josee and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ileana A Torres
- Division of Oncology, Department of Medicine, Stanford University, Stanford, CA, USA
| | - Eniola Oladipo
- Department of Radiation Oncology, Stanford University, Stanford, CA, USA
| | - Christin New
- Department of Orthopedic Surgery, Stanford University, Stanford, CA, USA
| | - Deborah E Kenney
- Department of Orthopedic Surgery, Stanford University, Stanford, CA, USA
| | - Raffi S Avedian
- Department of Orthopedic Surgery, Stanford University, Stanford, CA, USA
| | - Robert J Steffner
- Department of Orthopedic Surgery, Stanford University, Stanford, CA, USA
| | - Michael S Binkley
- Department of Radiation Oncology, Stanford University, Stanford, CA, USA
| | - David G Mohler
- Department of Orthopedic Surgery, Stanford University, Stanford, CA, USA
| | - William D Tap
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Weill Cornell Medical Center, New York, NY, USA
| | - Sandra P D'Angelo
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Weill Cornell Medical Center, New York, NY, USA
| | | | - Kristen N Ganjoo
- Division of Oncology, Department of Medicine, Stanford University, Stanford, CA, USA
| | - Nam Q Bui
- Division of Oncology, Department of Medicine, Stanford University, Stanford, CA, USA
| | | | - Aaron M Newman
- Department of Biomedical Data Science, Stanford University, Stanford, CA, USA
| | - Everett J Moding
- Department of Radiation Oncology, Stanford University, Stanford, CA, USA.
- Stanford Cancer Institute, Stanford University, Stanford, CA, USA.
| |
Collapse
|
7
|
Lee J, Cho S, Chung JH, Yoon SH, Shih BCH, Jung W, Jeon JH, Kim K, Jheon S. Prognosis of spread through air spaces in invasive mucinous lung adenocarcinoma after curative surgery. EUROPEAN JOURNAL OF SURGICAL ONCOLOGY 2024; 50:108053. [PMID: 38412587 DOI: 10.1016/j.ejso.2024.108053] [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: 12/26/2023] [Revised: 02/11/2024] [Accepted: 02/21/2024] [Indexed: 02/29/2024]
Abstract
INTRODUCTION The purpose of this study is to investigate the prognostic impact of spread through air spaces (STAS) in invasive mucinous adenocarcinoma (IMA). MATERIALS AND METHODS From 2015 to 2019, patients who underwent complete resection of IMA were extracted from the prospective database. Multivariable Cox-regression analysis and inverse probability of treatment weight (IPTW) - adjusted log-rank test for 5-year recurrence-free survival (RFS) were performed. RESULTS STAS was observed in 39.1% (53 out of 133). The STAS (+) group shows larger tumor size (2.9 ± 2.4 cm vs 3.8 ± 2.4 cm, p = 0.031) and higher incidence of lympho-vascular invasion (6 [7.5%] vs 18 [34.0%], p < 00.001) compared to the STAS (-) group. The 5-year RFS was 66.1% in the STAS (+) group and 91.8% in the STAS (-) group (p < 00.001), and the incidence of locoregional recurrence was significantly higher in the STAS (+) group than the STAS (-) group (1 [1.2%] vs 12 [22.6%], p < 00.001). Multivariable analysis revealed that STAS was associated with poor prognosis for all-recurrence (hazard ratio 2.81, 95% confidence interval 1.01-7.81, p = 0.048). After IPTW adjustment, 5-year RFS was 66.3% in the STAS (+) group and 92.9% in the STAS (-) group (p = 0.007), and risk for locoregional recurrence was greater in the STAS (+) group than the STAS (-) group (1.1 [0.9%] vs 20.8 [16.6%], p < 00.001). CONCLUSIONS STAS showed negative prognostic impact on all-recurrence, especially due to locoregional recurrence, after curative resection of IMA.
Collapse
Affiliation(s)
- Joonseok Lee
- Department of Thoracic and Cardiovascular Surgery, Seoul National University Bundang Hospital, Seongnam-si, Gyeonggi-do, Republic of Korea
| | - Sukki Cho
- Department of Thoracic and Cardiovascular Surgery, Seoul National University Bundang Hospital, Seongnam-si, Gyeonggi-do, Republic of Korea; Department of Thoracic and Cardiovascular Surgery, Seoul National University College of Medicine, Seoul, Republic of Korea.
| | - Jin-Haeng Chung
- Department of Pathology and Translational Medicine, Seoul National University Bundang Hospital, Seongnam-si, Gyeonggi-do, Republic of Korea; Department of Pathology, Seoul National University College of Medicine, Seoul, Republic of Korea.
| | - Seung Hwan Yoon
- Department of Thoracic and Cardiovascular Surgery, Seoul National University Bundang Hospital, Seongnam-si, Gyeonggi-do, Republic of Korea
| | - Beatrice Chia-Hui Shih
- Department of Thoracic and Cardiovascular Surgery, Seoul National University Bundang Hospital, Seongnam-si, Gyeonggi-do, Republic of Korea
| | - Woohyun Jung
- Department of Thoracic and Cardiovascular Surgery, Seoul National University Bundang Hospital, Seongnam-si, Gyeonggi-do, Republic of Korea
| | - Jae Hyun Jeon
- Department of Thoracic and Cardiovascular Surgery, Seoul National University Bundang Hospital, Seongnam-si, Gyeonggi-do, Republic of Korea
| | - Kwhanmien Kim
- Department of Thoracic and Cardiovascular Surgery, Seoul National University Bundang Hospital, Seongnam-si, Gyeonggi-do, Republic of Korea; Department of Thoracic and Cardiovascular Surgery, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Sanghoon Jheon
- Department of Thoracic and Cardiovascular Surgery, Seoul National University Bundang Hospital, Seongnam-si, Gyeonggi-do, Republic of Korea; Department of Thoracic and Cardiovascular Surgery, Seoul National University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
8
|
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
|
9
|
Silva D, Quintas C, Gonçalves J, Fresco P. β 2-Adrenoceptor Activation Favor Acquisition of Tumorigenic Properties in Non-Tumorigenic MCF-10A Breast Epithelial Cells. Cells 2024; 13:262. [PMID: 38334654 PMCID: PMC10854540 DOI: 10.3390/cells13030262] [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: 11/28/2023] [Revised: 01/22/2024] [Accepted: 01/26/2024] [Indexed: 02/10/2024] Open
Abstract
Noradrenaline and adrenaline, and their cognate receptors, are currently accepted to participate in cancer progression. They may also participate in cancer initiation, although their role in this phase is much less explored. The aim of this work was to study the influence of adrenergic stimulation in several processes related to breast cancer carcinogenesis, using several adrenergic agonists in the MCF-10A non-tumorigenic breast cells. Activation of the β-adrenoceptors promoted an epithelial phenotype in MCF-10A cells, revealed by an increased expression of the epithelial marker E-cadherin and a decrease in the mesenchymal markers, N-cadherin and vimentin. MCF-10A cell motility and migration were also impaired after the β-adrenoceptors activation. Concomitant with this effect, β-adrenoceptors decrease cell protrusions (lamellipodia and filopodia) while increasing cell adhesion. Activation of the β-adrenoceptors also decreases MCF-10A cell proliferation. When the MCF-10A cells were cultured under low attachment conditions, activation the of β- (likely β2) or of α2-adrenoceptors had protective effects against cell death, suggesting a pro-survival role of these adrenoceptors. Overall, our results showed that, in breast cells, adrenoceptor activation (mainly through β-adrenoceptors) may be a risk factor in breast cancer by inducing some cancer hallmarks, providing a mechanistic explanation for the increase in breast cancer incidences that may be associated with conditions that cause massive adrenergic stimulation, such as stress.
Collapse
Affiliation(s)
- Dany Silva
- Laboratory of Pharmacology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (D.S.); (C.Q.); (P.F.)
- UCIBIO—Applied Molecular Biosciences Unit, Associate Laboratory i4HB, Institute for Health and Bioeconomy, University of Porto, 4050-313 Porto, Portugal
| | - Clara Quintas
- Laboratory of Pharmacology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (D.S.); (C.Q.); (P.F.)
- UCIBIO—Applied Molecular Biosciences Unit, Associate Laboratory i4HB, Institute for Health and Bioeconomy, University of Porto, 4050-313 Porto, Portugal
| | - Jorge Gonçalves
- Laboratory of Pharmacology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (D.S.); (C.Q.); (P.F.)
- UCIBIO—Applied Molecular Biosciences Unit, Associate Laboratory i4HB, Institute for Health and Bioeconomy, University of Porto, 4050-313 Porto, Portugal
| | - Paula Fresco
- Laboratory of Pharmacology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (D.S.); (C.Q.); (P.F.)
- UCIBIO—Applied Molecular Biosciences Unit, Associate Laboratory i4HB, Institute for Health and Bioeconomy, University of Porto, 4050-313 Porto, Portugal
| |
Collapse
|
10
|
Jia S, Yang Y, Zhu Y, Yang W, Ling L, Wei Y, Fang X, Lin Q, Hamaï A, Mehrpour M, Gao J, Tan W, Xia Y, Chen J, Jiang W, Gong C. Association of FTH1-Expressing Circulating Tumor Cells With Efficacy of Neoadjuvant Chemotherapy for Patients With Breast Cancer: A Prospective Cohort Study. Oncologist 2024; 29:e25-e37. [PMID: 37390841 PMCID: PMC10769790 DOI: 10.1093/oncolo/oyad195] [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: 12/22/2022] [Accepted: 05/23/2023] [Indexed: 07/02/2023] Open
Abstract
BACKGROUND The association between different phenotypes and genotypes of circulating tumor cells (CTCs) and efficacy of neoadjuvant chemotherapy (NAC) remains uncertain. This study was conducted to evaluate the relationship of FTH1 gene-associated CTCs (F-CTC) with/without epithelial-mesenchymal transition (EMT) markers, or their dynamic changes with the efficacy of NAC in patients with non-metastatic breast cancer. PATIENTS AND METHODS This study enrolled 120 patients with non-metastatic breast cancer who planned to undergo NAC. The FTH1 gene and EMT markers in CTCs were detected before NAC (T0), after 2 cycles of chemotherapy (T1), and before surgery (T2). The associations of these different types of CTCs with rates of pathological complete response (pCR) and breast-conserving surgery (BCS) were evaluated using the binary logistic regression analysis. RESULTS F-CTC in peripheral blood ≥1 at T0 was an independent factor for pCR rate in patients with HER2-positive (odds ratio [OR]=0.08, 95% confidence interval [CI], 0.01-0.98, P = .048). The reduction in the number of F-CTC at T2 was an independent factor for BCS rate (OR = 4.54, 95% CI, 1.14-18.08, P = .03). CONCLUSIONS The number of F-CTC prior to NAC was related to poor response to NAC. Monitoring of F-CTC may help clinicians formulate personalized NAC regimens and implement BCS for patients with non-metastatic breast cancer.
Collapse
Affiliation(s)
- Shijie Jia
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People’s Republic of China
- Department of Breast Surgery, Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People’s Republic of China
| | - Yaping Yang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People’s Republic of China
- Department of Breast Surgery, Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People’s Republic of China
| | - Yingying Zhu
- Division of Clinical Research Design, Clinical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People’s Republic of China
| | - Wenqian Yang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People’s Republic of China
- Department of Breast Surgery, Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People’s Republic of China
| | - Li Ling
- Department of Medical Statistics, School of Public Health, Sun Yat-sen University, Guangzhou, People’s Republic of China
| | - Yanghui Wei
- The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, People’s Republic of China
| | - Xiaolin Fang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People’s Republic of China
- Department of Breast Surgery, Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People’s Republic of China
| | - Qun Lin
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People’s Republic of China
- Department of Breast Surgery, Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People’s Republic of China
| | - Ahmed Hamaï
- Institut Necker-Enfants Malades (INEM), Inserm U1151-CNRS UMR 8253, Université de Paris, Paris, France
| | - Maryam Mehrpour
- Institut Necker-Enfants Malades (INEM), Inserm U1151-CNRS UMR 8253, Université de Paris, Paris, France
| | - Jingbo Gao
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People’s Republic of China
- Department of Breast Surgery, Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People’s Republic of China
| | - Weige Tan
- Department of Breast Surgery, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, People’s Republic of China
| | - Yuan Xia
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People’s Republic of China
- Department of Breast Surgery, Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People’s Republic of China
| | - Jiayi Chen
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People’s Republic of China
- Department of Breast Surgery, Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People’s Republic of China
| | - Wenguo Jiang
- Cardiff China Medical Research Collaborative, Cardiff University School of Medicine, Cardiff University, Heath Park, Cardiff, UK
| | - Chang Gong
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People’s Republic of China
- Department of Breast Surgery, Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People’s Republic of China
| |
Collapse
|
11
|
Kwon MJ. Role of epithelial splicing regulatory protein 1 in cancer progression. Cancer Cell Int 2023; 23:331. [PMID: 38110955 PMCID: PMC10729575 DOI: 10.1186/s12935-023-03180-6] [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: 10/29/2023] [Accepted: 12/12/2023] [Indexed: 12/20/2023] Open
Abstract
As aberrant alternative splicing by either dysregulation or mutations of splicing factors contributes to cancer initiation and progression, splicing factors are emerging as potential therapeutic targets for cancer therapy. Therefore, pharmacological modulators targeting splicing factors have been under development. Epithelial splicing regulatory protein 1 (ESRP1) is an epithelial cell-specific splicing factor, whose downregulation is associated with epithelial-mesenchymal transition (EMT) by regulating alternative splicing of multiple genes, such as CD44, CTNND1, ENAH, and FGFR2. Consistent with the downregulation of ESRP1 during EMT, it has been initially revealed that high ESRP1 expression is associated with favorable prognosis and ESRP1 plays a tumor-suppressive role in cancer progression. However, ESRP1 has been found to promote cancer progression in some cancers, such as breast and ovarian cancers, indicating that it plays a dual role in cancer progression depending on the type of cancer. Furthermore, recent studies have reported that ESRP1 affects tumor growth by regulating the metabolism of tumor cells or immune cell infiltration in the tumor microenvironment, suggesting the novel roles of ESRP1 in addition to EMT. ESRP1 expression was also associated with response to anticancer drugs. This review describes current understanding of the roles and mechanisms of ESRP1 in cancer progression, and further discusses the emerging novel roles of ESRP1 in cancer and recent attempts to target splicing factors for cancer therapy.
Collapse
Affiliation(s)
- Mi Jeong Kwon
- Vessel-Organ Interaction Research Center (MRC), College of Pharmacy, Kyungpook National University, Daegu, Republic of Korea.
- BK21 FOUR KNU Community-Based Intelligent Novel Drug Discovery Education Unit, College of Pharmacy and Research Institute of Pharmaceutical Sciences, Kyungpook National University, 80 Daehak-ro, Buk-gu, Daegu, 41566, Republic of Korea.
| |
Collapse
|
12
|
Karacosta LG, Pancirer D, Preiss JS, Benson JA, Trope W, Shrager JB, Sung AW, Neal JW, Bendall SC, Wakelee H, Plevritis SK. Phenotyping EMT and MET cellular states in lung cancer patient liquid biopsies at a personalized level using mass cytometry. Sci Rep 2023; 13:21781. [PMID: 38065965 PMCID: PMC10709404 DOI: 10.1038/s41598-023-46458-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 11/01/2023] [Indexed: 12/18/2023] Open
Abstract
Malignant pleural effusions (MPEs) can be utilized as liquid biopsy for phenotyping malignant cells and for precision immunotherapy, yet MPEs are inadequately studied at the single-cell proteomic level. Here we leverage mass cytometry to interrogate immune and epithelial cellular profiles of primary tumors and pleural effusions (PEs) from early and late-stage non-small cell lung cancer (NSCLC) patients, with the goal of assessing epithelial-mesenchymal transition (EMT) and mesenchymal-epithelial transition (MET) states in patient specimens. By using the EMT-MET reference map PHENOSTAMP, we observe a variety of EMT states in cytokeratin positive (CK+) cells, and report for the first time MET-enriched CK+ cells in MPEs. We show that these states may be relevant to disease stage and therapy response. Furthermore, we found that the fraction of CD33+ myeloid cells in PEs was positively correlated to the fraction of CK+ cells. Longitudinal analysis of MPEs drawn 2 months apart from a patient undergoing therapy, revealed that CK+ cells acquired heterogeneous EMT features during treatment. We present this work as a feasibility study that justifies deeper characterization of EMT and MET states in malignant cells found in PEs as a promising clinical platform to better evaluate disease progression and treatment response at a personalized level.
Collapse
Affiliation(s)
- Loukia G Karacosta
- Department of Biomedical Data Science, Stanford University, Stanford, CA, 94305, USA
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, 77054, USA
| | - Danny Pancirer
- Stanford Cancer Institute - Clinical Trials Office, Stanford University, Stanford, CA, 94305, USA
| | - Jordan S Preiss
- Stanford Cancer Institute - Clinical Trials Office, Stanford University, Stanford, CA, 94305, USA
| | - Jalen A Benson
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA, 94305, USA
| | - Winston Trope
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA, 94305, USA
| | - Joseph B Shrager
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA, 94305, USA
- Palo Alto VA Health Care System, Palo Alto, USA
| | - Arthur Wai Sung
- Division of Pulmonary, Allergy & Critical Care Medicine, Department of Medicine, Stanford University, Stanford, CA, 94305, USA
| | - Joel W Neal
- Division of Oncology, Department of Medicine, Stanford University, Stanford, CA, 94305, USA
| | - Sean C Bendall
- Department of Pathology, Stanford University, Stanford, CA, 94305, USA
| | - Heather Wakelee
- Division of Oncology, Department of Medicine, Stanford University, Stanford, CA, 94305, USA
| | - Sylvia K Plevritis
- Department of Biomedical Data Science, Stanford University, Stanford, CA, 94305, USA.
- Department of Radiology, Stanford University, Stanford, CA, 94305, USA.
| |
Collapse
|
13
|
Tan W, Zhang Y, Wang J, Zheng Z, Xing L, Sun X. FDG PET/CT Tumor Dissemination Characteristic Predicts the Outcome of First-Line Systemic Therapy in Non-small Cell Lung Cancer. Acad Radiol 2023; 30:2904-2912. [PMID: 37202226 DOI: 10.1016/j.acra.2023.03.027] [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: 01/12/2023] [Revised: 03/19/2023] [Accepted: 03/20/2023] [Indexed: 05/20/2023]
Abstract
RATIONALE AND OBJECTIVES To explore the correlation between the tumor dissemination characteristic at 18F-fluoro-deoxyglucose positron emission tomography/computed tomography (18F-FDG PET/CT) images and the outcome of first-line systemic therapy for stage IV non-small cell lung cancer (NSCLC). MATERIALS AND METHODS The current retrospective study included 101 NSCLC patients receiving first-line systemic therapy with baseline 18F-FDG PET/CT images available. The distance between the two lesions that were the farthest apart was defined as Dmax to calculate the tumor dissemination. The tumor metabolic volume (MTV) of the primary tumor and the MTV of the whole-body tumor lesions (MTVwb) were calculated using 18F-FDG PET/CT imaging. The Kaplan-Meier survival analyses and Cox predictive model were performed to assess the relationship between the parameters and survival. RESULTS Dmax and MTVwb were independent prognostic factors for overall survival (OS) (p = 0.019 and p = 0.011, respectively) and progression-free survival (PFS) (p = 0.043 and p = 0.009, respectively). Poor PFS and OS were associated with high MTVwb (>54.0 cm3) and high Dmax (>48.5 cm) (p = 0.006 and p = 0.008, respectively). When MTVwb and Dmax were combined, three risk groups were stratified with no (score 0), one (score 1), or two (score 2) factors (p < 0.001 for PFS, p < 0.001 for OS). The group with a score of 0 had a considerably longer PFS and OS than those who received a score of 1 or 2 (PFS: 61.1%, 43.5%, and 21.1%, respectively, OS: 77.8%, 54.3%, and 36.8%, respectively). CONCLUSION The combination of tumor dissemination characteristic (Dmax) and tumor burden (MTVwb) can further improve the prognosis stratification of NSCLC.
Collapse
Affiliation(s)
- Weiyue Tan
- Department of Graduate, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China (W.T., Y.Z., J.W., Z.Z.); Department of Nuclear Medicine, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China (W.T., Y.Z., J.W., Z.Z., X.S.).
| | - Yi Zhang
- Department of Graduate, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China (W.T., Y.Z., J.W., Z.Z.); Department of Nuclear Medicine, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China (W.T., Y.Z., J.W., Z.Z., X.S.).
| | - Jie Wang
- Department of Graduate, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China (W.T., Y.Z., J.W., Z.Z.); Department of Nuclear Medicine, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China (W.T., Y.Z., J.W., Z.Z., X.S.).
| | - Zhonghang Zheng
- Department of Graduate, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China (W.T., Y.Z., J.W., Z.Z.); Department of Nuclear Medicine, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China (W.T., Y.Z., J.W., Z.Z., X.S.).
| | - Ligang Xing
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China (L.X.).
| | - Xiaorong Sun
- Department of Nuclear Medicine, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China (W.T., Y.Z., J.W., Z.Z., X.S.).
| |
Collapse
|
14
|
Lee JE, Park J, Kim EJ, Ko YH, Hong SA, Yang SH, Ahn YH. Noggin contributes to brain metastatic colonization of lung cancer cells. Cancer Cell Int 2023; 23:299. [PMID: 38012621 PMCID: PMC10683317 DOI: 10.1186/s12935-023-03155-7] [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: 07/12/2023] [Accepted: 11/21/2023] [Indexed: 11/29/2023] Open
Abstract
BACKGROUND Brain metastasis is a common complication among patients with lung cancer, yet the underlying mechanisms remain unclear. In this study, we aimed to investigate the pathogenesis of brain metastasis in lung cancer. METHODS We established highly colonizing metastatic lung cancer cells, A549-M2, through multiple implantations of A549 human lung cancer cells in the carotid artery of athymic nude mice. RESULTS Compared to parental cells (M0), M2 cells demonstrated slower growth in culture plates and soft agar, as well as lower motility and higher adhesion, key characteristics of mesenchymal-epithelial transition (MET). Further analysis revealed that M2 cells exhibited decreased expression of epithelial-mesenchymal transition markers, including ZEB1 and Vimentin. M2 cells also demonstrated reduced invasiveness in co-culture systems. RNA sequencing and gene set enrichment analysis confirmed that M2 cells underwent MET. Intriguingly, depletion of Noggin, a BMP antagonist, was observed in M2 cells, and replenishment of Noggin restored suppressed migration and invasion of M2 cells. In addition, Noggin knockdown in control M0 cells promoted cell attachment and suppressed cell migration, suggesting that Noggin reduction during brain colonization causes inhibition of migration and invasion of metastatic lung cancer cells. CONCLUSIONS Our results suggest that lung cancer cells undergo MET and lose their motility and invasiveness during brain metastatic colonization, which is dependent on Noggin.
Collapse
Affiliation(s)
- Jung Eun Lee
- Department of Neurosurgery, St. Vincent's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Jihye Park
- Department of Molecular Medicine and Inflammation-Cancer Microenvironment Research Center, College of Medicine, Ewha Womans University, 25 Magokdong-ro 2-gil, Gangseo-gu, Seoul, 07804, Republic of Korea
| | - Eun Ju Kim
- Department of Molecular Medicine and Inflammation-Cancer Microenvironment Research Center, College of Medicine, Ewha Womans University, 25 Magokdong-ro 2-gil, Gangseo-gu, Seoul, 07804, Republic of Korea
| | - Yoon Ho Ko
- Department of Internal Medicine, Division of Oncology, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Soon Auck Hong
- Department of Pathology, College of Medicine, Chung-Ang University, Seoul, Republic of Korea
| | - Seung Ho Yang
- Department of Neurosurgery, St. Vincent's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.
- Department of Neurosurgery, St. Vincent's Hospital, College of Medicine, The Catholic University of Korea, 93 Jungbu-daero, Paldal-gu, Suwon, 16247, Republic of Korea.
| | - Young-Ho Ahn
- Department of Molecular Medicine and Inflammation-Cancer Microenvironment Research Center, College of Medicine, Ewha Womans University, 25 Magokdong-ro 2-gil, Gangseo-gu, Seoul, 07804, Republic of Korea.
| |
Collapse
|
15
|
Voss G, Cassidy JR, Ceder Y. Functional consequences of A-to-I editing of miR-379 in prostate cancer cells. Sci Rep 2023; 13:16602. [PMID: 37789115 PMCID: PMC10547749 DOI: 10.1038/s41598-023-43775-7] [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: 03/22/2023] [Accepted: 09/28/2023] [Indexed: 10/05/2023] Open
Abstract
Prostate cancer is the predominant cause of cancer in men, but there is still a lack of biomarkers and treatments for metastatic spread. The initial promise of microRNAs to provide avenues to solve these problems has been dampened by the realisation that microRNAs co-exist in multiple functionally distinct isoforms, for example due to A-to-I editing. We recently found that A-to-I-editing of microRNA-379 (miR-379) was associated with prostate cancer, and that only the unedited isoform was negatively correlated with aggressive disease. Here, we set out to decipher the biological effects of unedited and edited miR-379 in prostate cancer cells. After transfection of four different prostate cancer cell lines with isoform-specific miR-379 mimics, we performed assays for cell growth, colony formation, migration, cell-cell adhesion, and analysed epithelial-mesenchymal transition (EMT) and stemness markers. We found that unedited miR-379 affected cell growth, with a promoting function in androgen receptor (AR)-negative cells and an inhibiting effect in AR-positive cells. This is supported by our in silico analysis that found unedited miR-379 targets are predicted to be predominantly involved in cellular proliferation whereas the targets of edited miR-379 are not. We further found that both miR-379 isoforms could promote colony formation, migration, and cell-cell adhesion. Overall, our data suggests that editing of miR-379 attenuates the growth-suppressive function of unedited miR-379 in androgen-sensitive prostate cancer cells, thereby promoting tumor growth.
Collapse
Affiliation(s)
- Gjendine Voss
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - James R Cassidy
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Yvonne Ceder
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, Lund, Sweden.
| |
Collapse
|
16
|
Valakos D, Klagkou E, Kokkalis A, Polyzos A, Kyrilis FL, Banos A, Vatsellas G, Pliatska M, Ford E, Stravopodis DJ, Thanos D. Combinatorial targeting of a specific EMT/MET network by macroH2A variants safeguards mesenchymal identity. PLoS One 2023; 18:e0288005. [PMID: 37432970 DOI: 10.1371/journal.pone.0288005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 06/16/2023] [Indexed: 07/13/2023] Open
Abstract
Generation of induced pluripotent stem cells from specialized cell types provides an excellent model to study how cells maintain their stability, and how they can change identity, especially in the context of disease. Previous studies have shown that chromatin safeguards cell identity by acting as a barrier to reprogramming. We investigated mechanisms by which the histone macroH2A variants inhibit reprogramming and discovered that they work as gate keepers of the mesenchymal cell state by blocking epithelial transition, a step required for reprogramming of mouse fibroblasts. More specifically, we found that individual macroH2A variants regulate the expression of defined sets of genes, whose overall function is to stabilize the mesenchymal gene expression program, thus resisting reprogramming. We identified a novel gene network (MSCN, mesenchymal network) composed of 63 macroH2A-regulated genes related to extracellular matrix, cell membrane, signaling and the transcriptional regulators Id2 and Snai2, all of which function as guardians of the mesenchymal phenotype. ChIP-seq and KD experiments revealed a macroH2A variant-specific combinatorial targeting of the genes reconstructing the MSCN, thus generating robustness in gene expression programs to resist cellular reprogramming.
Collapse
Affiliation(s)
- Dimitrios Valakos
- Biomedical Research Foundation, Academy of Athens, Athens, Greece
- Section of Biochemistry and Molecular Biology, Department of Biology, School of Science, National and Kapodistrian University of Athens (NKUA), Zografou, Athens, Greece
| | - Eleftheria Klagkou
- Biomedical Research Foundation, Academy of Athens, Athens, Greece
- Section of Biochemistry and Molecular Biology, Department of Biology, School of Science, National and Kapodistrian University of Athens (NKUA), Zografou, Athens, Greece
| | - Antonis Kokkalis
- Biomedical Research Foundation, Academy of Athens, Athens, Greece
| | | | - Fotis L Kyrilis
- Biomedical Research Foundation, Academy of Athens, Athens, Greece
| | - Aggelos Banos
- Biomedical Research Foundation, Academy of Athens, Athens, Greece
| | | | - Maria Pliatska
- Biomedical Research Foundation, Academy of Athens, Athens, Greece
| | - Ethan Ford
- Biomedical Research Foundation, Academy of Athens, Athens, Greece
| | - Dimitrios J Stravopodis
- Section of Cell Biology and Biophysics, Department of Biology, School of Science, National and Kapodistrian University of Athens (NKUA), Zografou, Athens, Greece
| | - Dimitris Thanos
- Biomedical Research Foundation, Academy of Athens, Athens, Greece
| |
Collapse
|
17
|
Arena GO, Forte S, Abdouh M, Vanier C, Corbeil D, Lorico A. Horizontal Transfer of Malignant Traits and the Involvement of Extracellular Vesicles in Metastasis. Cells 2023; 12:1566. [PMID: 37371036 PMCID: PMC10297028 DOI: 10.3390/cells12121566] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 05/31/2023] [Accepted: 06/02/2023] [Indexed: 06/29/2023] Open
Abstract
Metastases are responsible for the vast majority of cancer deaths, yet most therapeutic efforts have focused on targeting and interrupting tumor growth rather than impairing the metastatic process. Traditionally, cancer metastasis is attributed to the dissemination of neoplastic cells from the primary tumor to distant organs through blood and lymphatic circulation. A thorough understanding of the metastatic process is essential to develop new therapeutic strategies that improve cancer survival. Since Paget's original description of the "Seed and Soil" hypothesis over a hundred years ago, alternative theories and new players have been proposed. In particular, the role of extracellular vesicles (EVs) released by cancer cells and their uptake by neighboring cells or at distinct anatomical sites has been explored. Here, we will outline and discuss these alternative theories and emphasize the horizontal transfer of EV-associated biomolecules as a possibly major event leading to cell transformation and the induction of metastases. We will also highlight the recently discovered intracellular pathway used by EVs to deliver their cargoes into the nucleus of recipient cells, which is a potential target for novel anti-metastatic strategies.
Collapse
Affiliation(s)
- Goffredo O. Arena
- Department of Surgery, McGill University, Montréal, QC H3A 0G4, Canada;
- Fondazione Istituto G. Giglio, 90015 Cefalù, Italy
- Mediterranean Institute of Oncology, 95029 Viagrande, Italy;
| | - Stefano Forte
- Mediterranean Institute of Oncology, 95029 Viagrande, Italy;
| | - Mohamed Abdouh
- Cancer Research Program, Research Institute, McGill University Health Centre, Montréal, QC H3A 0G4, Canada;
| | - Cheryl Vanier
- Touro University Nevada College of Medicine, Henderson, NV 89014, USA;
| | - Denis Corbeil
- Biotechnology Center (BIOTEC) and Center for Molecular and Cellular Bioengineering, Technische Universität Dresden, 01307 Dresden, Germany;
| | - Aurelio Lorico
- Mediterranean Institute of Oncology, 95029 Viagrande, Italy;
- Touro University Nevada College of Medicine, Henderson, NV 89014, USA;
| |
Collapse
|
18
|
Ruscone M, Montagud A, Chavrier P, Destaing O, Bonnet I, Zinovyev A, Barillot E, Noël V, Calzone L. Multiscale model of the different modes of cancer cell invasion. Bioinformatics 2023; 39:btad374. [PMID: 37289551 PMCID: PMC10293590 DOI: 10.1093/bioinformatics/btad374] [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: 11/14/2022] [Revised: 05/25/2023] [Accepted: 06/07/2023] [Indexed: 06/10/2023] Open
Abstract
MOTIVATION Mathematical models of biological processes altered in cancer are built using the knowledge of complex networks of signaling pathways, detailing the molecular regulations inside different cell types, such as tumor cells, immune and other stromal cells. If these models mainly focus on intracellular information, they often omit a description of the spatial organization among cells and their interactions, and with the tumoral microenvironment. RESULTS We present here a model of tumor cell invasion simulated with PhysiBoSS, a multiscale framework, which combines agent-based modeling and continuous time Markov processes applied on Boolean network models. With this model, we aim to study the different modes of cell migration and to predict means to block it by considering not only spatial information obtained from the agent-based simulation but also intracellular regulation obtained from the Boolean model. Our multiscale model integrates the impact of gene mutations with the perturbation of the environmental conditions and allows the visualization of the results with 2D and 3D representations. The model successfully reproduces single and collective migration processes and is validated on published experiments on cell invasion. In silico experiments are suggested to search for possible targets that can block the more invasive tumoral phenotypes. AVAILABILITY AND IMPLEMENTATION https://github.com/sysbio-curie/Invasion_model_PhysiBoSS.
Collapse
Affiliation(s)
- Marco Ruscone
- Institut Curie, Université PSL, F-75005 Paris, France
- INSERM, U900, F-75005 Paris, France
- Mines ParisTech, Université PSL, F-75005 Paris, France
- Sorbonne Université, Collège Doctoral, F-75005 Paris, France
| | | | - Philippe Chavrier
- Institut Curie, PSL Research University, CNRS, UMR 144, Paris, France
| | - Olivier Destaing
- Institute for Advanced Biosciences, Centre de Recherche Université Grenoble Alpes, Inserm U 1209, CNRS UMR 5309, France
| | - Isabelle Bonnet
- Institut Curie, Université PSL, Sorbonne Université, CNRS UMR168, Laboratoire Physico Chimie Curie, Paris, France
| | - Andrei Zinovyev
- Institut Curie, Université PSL, F-75005 Paris, France
- INSERM, U900, F-75005 Paris, France
- Mines ParisTech, Université PSL, F-75005 Paris, France
| | - Emmanuel Barillot
- Institut Curie, Université PSL, F-75005 Paris, France
- INSERM, U900, F-75005 Paris, France
- Mines ParisTech, Université PSL, F-75005 Paris, France
| | - Vincent Noël
- Institut Curie, Université PSL, F-75005 Paris, France
- INSERM, U900, F-75005 Paris, France
- Mines ParisTech, Université PSL, F-75005 Paris, France
| | - Laurence Calzone
- Institut Curie, Université PSL, F-75005 Paris, France
- INSERM, U900, F-75005 Paris, France
- Mines ParisTech, Université PSL, F-75005 Paris, France
| |
Collapse
|
19
|
O'Connell AE, Raveenthiraraj S, Adegboye C, Qi W, Khetani RS, Singh A, Sundaram N, Emeonye C, Lin J, Goldsmith JD, Thiagarajah JR, Carlone DL, Turner JR, Agrawal PB, Helmrath M, Breault DT. WNT2B Deficiency Causes Increased Susceptibility to Colitis in Mice and Impairs Intestinal Epithelial Development in Humans. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.21.537894. [PMID: 37131772 PMCID: PMC10153278 DOI: 10.1101/2023.04.21.537894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Background and aims WNT2B is a canonical Wnt ligand previously thought to be fully redundant with other Wnts in the intestinal epithelium. However, humans with WNT2B deficiency have severe intestinal disease, highlighting a critical role for WNT2B. We sought to understand how WNT2B contributes to intestinal homeostasis. Methods We investigated the intestinal health of Wnt2b knock out (KO) mice. We assessed the impact of inflammatory challenge to the small intestine, using anti-CD3χ antibody, and to the colon, using dextran sodium sulfate (DSS). In addition, we generated human intestinal organoids (HIOs) from WNT2B-deficient human iPSCs for transcriptional and histological analyses. Results Mice with WNT2B deficiency had significantly decreased Lgr5 expression in the small intestine and profoundly decreased expression in the colon, but normal baseline histology. The small intestinal response to anti-CD3χ antibody was similar in Wnt2b KO and wild type (WT) mice. In contrast, the colonic response to DSS in Wnt2b KO mice showed an accelerated rate of injury, featuring earlier immune cell infiltration and loss of differentiated epithelium compared to WT. WNT2B-deficient HIOs showed abnormal epithelial organization and an increased mesenchymal gene signature. Conclusion WNT2B contributes to maintenance of the intestinal stem cell pool in mice and humans. WNT2B deficient mice, which do not have a developmental phenotype, show increased susceptibility to colonic injury but not small intestinal injury, potentially due to a higher reliance on WNT2B in the colon compared to the small intestine.WNT2B deficiency causes a developmental phenotype in human intestine with HIOs showing a decrease in their mesenchymal component and WNT2B-deficient patients showing epithelial disorganization. Data Transparency Statement All RNA-Seq data will be available through online repository as indicated in Transcript profiling. Any other data will be made available upon request by emailing the study authors.
Collapse
|
20
|
El Hadi C, Hilal G, Aoun R. Enhancing cancer treatment and understanding through clustering of gene responses to categorical stressors. Sci Rep 2023; 13:6517. [PMID: 37085609 PMCID: PMC10121664 DOI: 10.1038/s41598-023-33785-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 04/19/2023] [Indexed: 04/23/2023] Open
Abstract
Cancer cells have a unique metabolic activity in the glycolysis pathway compared to normal cells, which allows them to maintain their growth and proliferation. Therefore, inhibition of glycolytic pathways may be a promising therapeutic approach for cancer treatment. In this novel study, we analyzed the genetic responses of cancer cells to stressors, particularly to drugs that target the glycolysis pathway. Gene expression data for experiments on different cancer cell types were extracted from the Gene Expression Omnibus and the expression fold change was then clustered after dimensionality reduction. We identified four groups of responses: the first and third were most affected by anti-glycolytic drugs, especially those acting on multiple pathways at once, and consisted mainly of squamous and mesenchymal tissues, showing higher mitotic inhibition and apoptosis. The second and fourth groups were relatively unaffected by treatment, comprising mainly gynecologic and hormone-sensitive groups, succumbing least to glycolysis inhibitors. Hexokinase-targeted drugs mainly showed this blunted effect on cancer cells. This study highlights the importance of analyzing the molecular states of cancer cells to identify potential targets for personalized cancer therapies and to improve our understanding of the disease.
Collapse
Affiliation(s)
| | - George Hilal
- Cancer and Metabolism Laboratory, Faculty of Medicine, Saint-Joseph University, Beirut, Lebanon
| | - Rita Aoun
- Cancer and Metabolism Laboratory, Faculty of Medicine, Saint-Joseph University, Beirut, Lebanon
| |
Collapse
|
21
|
Monti N, Dinicola S, Querqui A, Fabrizi G, Fedeli V, Gesualdi L, Catizone A, Unfer V, Bizzarri M. Myo-Inositol Reverses TGF-β1-Induced EMT in MCF-10A Non-Tumorigenic Breast Cells. Cancers (Basel) 2023; 15:cancers15082317. [PMID: 37190245 DOI: 10.3390/cancers15082317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 04/12/2023] [Accepted: 04/13/2023] [Indexed: 05/17/2023] Open
Abstract
Epithelial-Mesenchymal Transition (EMT), triggered by external and internal cues in several physiological and pathological conditions, elicits the transformation of epithelial cells into a mesenchymal-like phenotype. During EMT, epithelial cells lose cell-to-cell contact and acquire unusual motility/invasive capabilities. The associated architectural and functional changes destabilize the epithelial layer consistency, allowing cells to migrate and invade the surrounding tissues. EMT is a critical step in the progression of inflammation and cancer, often sustained by a main driving factor as the transforming growth factor-β1 (TGF-β1). Antagonizing EMT has recently gained momentum as an attractive issue in cancer treatment and metastasis prevention. Herein, we demonstrate the capability of myo-inositol (myo-Ins) to revert the EMT process induced by TGF-β1 on MCF-10A breast cells. Upon TGF-β1 addition, cells underwent a dramatic phenotypic transformation, as witnessed by structural (disappearance of the E-cadherin-β-catenin complexes and the emergence of a mesenchymal shape) and molecular modifications (increase in N-cadherin, Snai1, and vimentin), including the release of increased collagen and fibronectin. However, following myo-Ins, those changes were almost completely reverted. Inositol promotes the reconstitution of E-cadherin-β-catenin complexes, decreasing the expression of genes involved in EMT, while promoting the re-expression of epithelial genes (keratin-18 and E-cadherin). Noticeably, myo-Ins efficiently inhibits the invasiveness and migrating capability of TGF-β1 treated cells, also reducing the release of metalloproteinase (MMP-9) altogether with collagen synthesis, allowing for the re-establishment of appropriate cell-to-cell junctions, ultimately leading the cell layer back towards a more compact state. Inositol effects were nullified by previous treatment with an siRNA construct to inhibit CDH1 transcripts and, hence, E-cadherin synthesis. This finding suggests that the reconstitution of E-cadherin complexes is an irreplaceable step in the inositol-induced reversion of EMT. Overall, such a result advocates for the useful role of myo-Ins in cancer treatment.
Collapse
Affiliation(s)
- Noemi Monti
- Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161 Rome, Italy
- Systems Biology Group Laboratory, Sapienza University, 00161 Rome, Italy
| | - Simona Dinicola
- Systems Biology Group Laboratory, Sapienza University, 00161 Rome, Italy
| | - Alessandro Querqui
- Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161 Rome, Italy
- Systems Biology Group Laboratory, Sapienza University, 00161 Rome, Italy
| | - Gianmarco Fabrizi
- Systems Biology Group Laboratory, Sapienza University, 00161 Rome, Italy
| | - Valeria Fedeli
- Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161 Rome, Italy
- Systems Biology Group Laboratory, Sapienza University, 00161 Rome, Italy
| | - Luisa Gesualdi
- Section of Histology and Embryology, Department of Anatomy, Histology, Forensic Medicine and Orthopedics, Sapienza University of Rome, Viale Regina Elena 336, 00161 Rome, Italy
| | - Angela Catizone
- Section of Histology and Embryology, Department of Anatomy, Histology, Forensic Medicine and Orthopedics, Sapienza University of Rome, Viale Regina Elena 336, 00161 Rome, Italy
| | - Vittorio Unfer
- Systems Biology Group Laboratory, Sapienza University, 00161 Rome, Italy
- The Experts Group on Inositol in Basic and Clinical Research (EGOI), 00161 Rome, Italy
- Gynecology Department, UniCamillus-Saint Camillus International University of Health and Medical Sciences, 00161 Rome, Italy
| | - Mariano Bizzarri
- Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161 Rome, Italy
- Systems Biology Group Laboratory, Sapienza University, 00161 Rome, Italy
- The Experts Group on Inositol in Basic and Clinical Research (EGOI), 00161 Rome, Italy
| |
Collapse
|
22
|
Abboud Asleh M, Zaher M, Asleh J, Jadon J, Shaulov L, Yelin R, Schultheiss TM. A morphogenetic wave in the chick embryo lateral mesoderm generates mesenchymal-epithelial transition through a 3D-rosette intermediate. Dev Cell 2023:S1534-5807(23)00133-8. [PMID: 37080204 DOI: 10.1016/j.devcel.2023.03.017] [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: 07/19/2022] [Revised: 02/24/2023] [Accepted: 03/24/2023] [Indexed: 04/22/2023]
Abstract
Formation of epithelia through mesenchymal-epithelial transition (MET) is essential for embryonic development and for many physiological and pathological processes. This study investigates MET in vivo in the chick embryo lateral mesoderm, where a multilayered mesenchyme transforms into two parallel epithelial sheets that constitute the coelomic lining of the embryonic body cavity. Prior to MET initiation, mesenchymal cells exhibit non-polarized distribution of multiple polarity markers, albeit not aPKC. We identified an epithelializing wave that sweeps across the lateral mesoderm, the wavefront of which is characterized by the accumulation of basal fibronectin and a network of 3D rosettes composed of polarized, wedge-shaped cells surrounding a central focus of apical markers, now including aPKC. Initiation of the MET process is dependent on extracellular matrix-integrin signaling acting through focal adhesion kinase and talin, whereas progression through the rosette phase requires aPKC function. We present a stepwise model for MET, comprising polarization, 3D-rosette, and epithelialization stages.
Collapse
Affiliation(s)
- Manar Abboud Asleh
- Department of Genetics and Developmental Biology, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 31096, Israel
| | - Mira Zaher
- Department of Genetics and Developmental Biology, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 31096, Israel
| | - Jad Asleh
- Department of Genetics and Developmental Biology, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 31096, Israel
| | - Julian Jadon
- Department of Genetics and Developmental Biology, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 31096, Israel
| | - Lihi Shaulov
- Department of Genetics and Developmental Biology, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 31096, Israel
| | - Ronit Yelin
- Department of Genetics and Developmental Biology, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 31096, Israel
| | - Thomas M Schultheiss
- Department of Genetics and Developmental Biology, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 31096, Israel.
| |
Collapse
|
23
|
Li D, Xia L, Huang P, Wang Z, Guo Q, Huang C, Leng W, Qin S. Heterogeneity and plasticity of epithelial-mesenchymal transition (EMT) in cancer metastasis: Focusing on partial EMT and regulatory mechanisms. Cell Prolif 2023:e13423. [PMID: 36808651 DOI: 10.1111/cpr.13423] [Citation(s) in RCA: 30] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 01/05/2023] [Accepted: 01/27/2023] [Indexed: 02/22/2023] Open
Abstract
Epithelial-mesenchymal transition (EMT) or mesenchymal-epithelial transition (MET) plays critical roles in cancer metastasis. Recent studies, especially those based on single-cell sequencing, have revealed that EMT is not a binary process, but a heterogeneous and dynamic disposition with intermediary or partial EMT states. Multiple double-negative feedback loops involved by EMT-related transcription factors (EMT-TFs) have been identified. These feedback loops between EMT drivers and MET drivers finely regulate the EMT transition state of the cell. In this review, the general characteristics, biomarkers and molecular mechanisms of different EMT transition states were summarized. We additionally discussed the direct and indirect roles of EMT transition state in tumour metastasis. More importantly, this article provides direct evidence that the heterogeneity of EMT is closely related to the poor prognosis in gastric cancer. Notably, a seesaw model was proposed to explain how tumour cells regulate themselves to remain in specific EMT transition states, including epithelial state, hybrid/intermediate state and mesenchymal state. Additionally, this article also provides a review of the current status, limitations and future perspectives of EMT signalling in clinical applications.
Collapse
Affiliation(s)
- Dandan Li
- Department of Stomatology, Taihe Hospital and Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, China.,Laboratory of Tumor Biology, Academy of Bio-medicine Research, Hubei University of Medicine, Shiyan, China
| | - Lingyun Xia
- Department of Stomatology, Taihe Hospital and Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, China
| | - Pan Huang
- Laboratory of Tumor Biology, Academy of Bio-medicine Research, Hubei University of Medicine, Shiyan, China
| | - Zidi Wang
- Laboratory of Tumor Biology, Academy of Bio-medicine Research, Hubei University of Medicine, Shiyan, China
| | - Qiwei Guo
- Laboratory of Tumor Biology, Academy of Bio-medicine Research, Hubei University of Medicine, Shiyan, China
| | - Congcong Huang
- Laboratory of Tumor Biology, Academy of Bio-medicine Research, Hubei University of Medicine, Shiyan, China
| | - Weidong Leng
- Department of Stomatology, Taihe Hospital and Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, China
| | - Shanshan Qin
- Department of Stomatology, Taihe Hospital and Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, China.,Laboratory of Tumor Biology, Academy of Bio-medicine Research, Hubei University of Medicine, Shiyan, China
| |
Collapse
|
24
|
Lin YP, Hseu YC, Thiyagarajan V, Vadivalagan C, Pandey S, Lin KY, Hsu YT, Liao JW, Lee CC, Yang HL. The in vitro and in vivo anticancer activities of Antrodia salmonea through inhibition of metastasis and induction of ROS-mediated apoptotic and autophagic cell death in human glioblastoma cells. Biomed Pharmacother 2023; 158:114178. [PMID: 36916401 DOI: 10.1016/j.biopha.2022.114178] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 12/27/2022] [Accepted: 12/28/2022] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Antrodia salmonea (AS) exhibits anticancer activities against various cancers. OBJECTIVE This study investigated the anticancer activities of AS on human glioblastoma (GBM8401 and U87MG) cells both in vitro and in vivo and explained the underlying molecular mechanism. METHODS MTT, colony formation, migration/invasion assay, immunoblotting, immunofluorescence, TUNEL, Annexin V/PI staining, AO staining, GFP-LC3 transfection, TEM, qPCR, siLC3, DCFH2-DA assay, and xenografted-nude mice were used to assess the potential of AS therapy. RESULTS AS treatment retarded growth and suppressed colony formation in glioblastoma cells. AS attenuates EMT by suppressing invasion and migration, increasing E-cadherin expression, decreasing Twist, Snail, and N-cadherin expression, and inhibiting Wnt/β-catenin pathways in GBM8401 and U87MG cells. Furthermore, AS induced apoptosis by activating caspase-3, cleaving PARP, and dysregulating Bax and Bcl-2 in both cell lines. TUNEL assay and Annexin V/PI staining indicated AS-mediated late apoptosis. Interestingly, AS induced autophagic cell death by LC3-II accumulation, AVO formation, autophagosome GFP-LC3 puncta, p62/SQSTM1 expression, and ATG4B inhibition in GBM8401 and U87MG cells. TEM data revealed that AS favored autophagosome and autolysosome formation. The autophagy inhibitors 3-MA/CQ and LC3 knockdown suppressed AS-induced apoptosis in glioblastoma cells, indicating that the inhibition of autophagy decreased AS-induced apoptosis. Notably, the antioxidant N-acetylcysteine (NAC) inhibited AS-mediated ROS production and AS-induced apoptotic and autophagic cell death. Furthermore, AS induced ROS-mediated inhibition of the PI3K/AKT/mTOR signaling pathway. AS reduced the tumor burden in GBM8401-xenografted nude mice and significantly modulated tumor xenografts by inducing anti-EMT, apoptosis, and autophagy. AS could be a potential antitumor agent in human glioblastoma treatment.
Collapse
Affiliation(s)
- Yi-Pin Lin
- Institute of Nutrition, College of Health Care, China Medical University, Taichung 40402, Taiwan, ROC
| | - You-Cheng Hseu
- Department of Cosmeceutics, College of Pharmacy, China Medical University, Taichung 40402, Taiwan, ROC; Department of Health and Nutrition Biotechnology, Asia University, Taichung 41354, Taiwan, ROC; Chinese Medicine Research Center, China Medical University, Taichung 40402, Taiwan, ROC; Research Center of Chinese Herbal Medicine, China Medical University, Taichung 40402, Taiwan, ROC.
| | - Varadharajan Thiyagarajan
- Department of Cosmeceutics, College of Pharmacy, China Medical University, Taichung 40402, Taiwan, ROC
| | - Chithravel Vadivalagan
- Department of Cosmeceutics, College of Pharmacy, China Medical University, Taichung 40402, Taiwan, ROC
| | - Sudhir Pandey
- Department of Pharmacology, Faculty of Pharmacy, Mahidol University, Bangkok 10400, Thailand
| | - Kai-Yuan Lin
- Department of Medical Research, Chi-Mei Medical Center, Tainan 710, Taiwan, ROC; Department of Biotechnology, Chia Nan University of Pharmacy and Science, Tainan 71710, Taiwan, ROC
| | - Yuan-Tai Hsu
- Institute of Nutrition, College of Health Care, China Medical University, Taichung 40402, Taiwan, ROC
| | - Jiunn-Wang Liao
- Graduate Institute of Veterinary Pathology, National Chung-Hsing University, Taichung 402, Taiwan, ROC
| | - Chuan-Chen Lee
- Department of Health and Nutrition Biotechnology, Asia University, Taichung 41354, Taiwan, ROC
| | - Hsin-Ling Yang
- Institute of Nutrition, College of Health Care, China Medical University, Taichung 40402, Taiwan, ROC.
| |
Collapse
|
25
|
Zhu Y, Yin WF, Yu P, Zhang C, Sun MH, Kong LY, Yang L. Meso-Hannokinol inhibits breast cancer bone metastasis via the ROS/JNK/ZEB1 axis. Phytother Res 2023. [PMID: 36726293 DOI: 10.1002/ptr.7732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 06/25/2022] [Accepted: 07/11/2022] [Indexed: 02/03/2023]
Abstract
Distal metastases from breast cancer, especially bone metastases, are extremely common in the late stages of the disease and are associated with a poor prognosis. EMT is a biomarker of the early process of bone metastasis, and MMP-9 and MMP-13 are important osteoclastic activators. Previously, we found that meso-Hannokinol (HA) could significantly inhibit EMT and MMP-9 and MMP-13 expressions in breast cancer cells. On this basis, we further explored the role of HA in breast cancer bone metastasis. In vivo, we established a breast cancer bone metastasis model by intracardially injecting breast cancer cells. Intraperitoneal injections of HA significantly reduced breast cancer cell metastasis to the leg bone in mice and osteolytic lesions caused by breast cancer. In vitro, HA inhibited the migration and invasion of breast cancer cells and suppressed the expressions of EMT, MMP-9, MMP-13, and other osteoclastic activators. HA inhibited EMT and MMP-9 by activating the ROS/JNK pathway as demonstrated by siJNK and SP600125 inhibition of JNK phosphorylation and NAC scavenging of ROS accumulation. Moreover, HA promoted bone formation and inhibited bone resorption in vitro. In conclusion, our findings suggest that HA may be an excellent candidate for treating breast cancer bone metastasis.
Collapse
Affiliation(s)
- Yuan Zhu
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Wei-Feng Yin
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Pei Yu
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Chao Zhang
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Ming-Hui Sun
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, People's Republic of China
| | - Ling-Yi Kong
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Lei Yang
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, People's Republic of China
| |
Collapse
|
26
|
"Pulsed Hypoxia" Gradually Reprograms Breast Cancer Fibroblasts into Pro-Tumorigenic Cells via Mesenchymal-Epithelial Transition. Int J Mol Sci 2023; 24:ijms24032494. [PMID: 36768815 PMCID: PMC9916667 DOI: 10.3390/ijms24032494] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 01/24/2023] [Accepted: 01/24/2023] [Indexed: 01/31/2023] Open
Abstract
Hypoxia arises in most growing solid tumors and can lead to pleotropic effects that potentially increase tumor aggressiveness and resistance to therapy through regulation of the expression of genes associated with the epithelial-mesenchymal transition (EMT) and mesenchymal-epithelial transition (MET). The main goal of the current work was to obtain and investigate the intermediate phenotype of tumor cells undergoing the hypoxia-dependent transition from fibroblast to epithelial morphology. Primary breast cancer fibroblasts BrC4f, being cancer-associated fibroblasts, were subjected to one or two rounds of "pulsed hypoxia" (PH). PH induced transformation of fibroblast-shaped cells to semi-epithelial cells. Western blot analysis, fluorescent microscopy and flow cytometry of transformed cells demonstrated the decrease in the mesenchymal markers vimentin and N-cad and an increase in the epithelial marker E-cad. These cells kept mesenchymal markers αSMA and S100A4 and high ALDH activity. Real-time PCR data of the cells after one (BrC4f_Hyp1) and two (BrC4f_Hyp2) rounds of PH showed consistent up-regulation of TWIST1 gene as an early response and ZEB1/2 and SLUG transcriptional activity as a subsequent response. Reversion of BrC4f_Hyp2 cells to normoxia conditions converted them to epithelial-like cells (BrC4e) with decreased expression of EMT genes and up-regulation of MET-related OVOL2 and c-MYC genes. Transplantation of BrC4f and BrC4f_Hyp2 cells into SCID mice showed the acceleration of tumor growth up to 61.6% for BrC4f_Hyp2 cells. To summarize, rounds of PH imitate the MET process of tumorigenesis in which cancer-associated fibroblasts pass through intermediate stages and become more aggressive epithelial-like tumor cells.
Collapse
|
27
|
Wang Q, Liao C, Tan Z, Li X, Guan X, Li H, Tian Z, Liu J, An J. FUT6 inhibits the proliferation, migration, invasion, and EGF-induced EMT of head and neck squamous cell carcinoma (HNSCC) by regulating EGFR/ERK/STAT signaling pathway. Cancer Gene Ther 2023; 30:182-191. [PMID: 36151332 DOI: 10.1038/s41417-022-00530-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 08/10/2022] [Accepted: 08/26/2022] [Indexed: 02/07/2023]
Abstract
Glycosylation change is one of the landmark events of tumor occurrence and development, and tumor cells may be inhibited by regulating the aberrant expression of glycosyltransferases. Currently, fucosyltransferase VI (FUT6), which is involved in the synthesis of α-1, 3 fucosyl bond, has been detected to be closely associated with multiple tumors, but its function and mechanism in head and neck squamous cell carcinoma (HNSCC) still need further research. In this study, FUT6 knockdown and overexpression strategies were used to investigate the effects of FUT6 on cell proliferation, migration, and invasion, as well as the growth and metastasis of HNSCC in a xenografts mouse model. The protein expression levels of epidermal growth factor receptor (EGFR), extracellular signal-regulated kinase (ERK), Signal Transducer and Activator of Transcription (STAT), protein kinase B (AKT), c-Myc, and epithelial-mesenchymal transition (EMT) markers were determined by western blot analysis. Our research found that the mRNA expression of FUT6 was lower in HNSCC tissues than in normal mucosal epithelial tissues. In Cal-27 and FaDu cells, FUT6 overexpression inhibited cell proliferation, migration and invasion, causing upregulation of ZO-1 and E-cadherin, downregulation of N-cadherin and Vimentin, and finally decreased the phosphorylation levels of EGFR, ERK, STAT, and c-Myc. In HSC-3 cells, knockdown of FUT6 promoted cell proliferation, migration and invasion, downregulating ZO-1 and E-cadherin, upregulating N-cadherin and Vimentin, and increased the phosphorylation levels of EGFR, ERK, STAT, and c-Myc. In the HNSCC xenografts mouse, FUT6 overexpression inhibited tumor growth and metastasis. In summary, FUT6 controls the proliferation, migration, invasion, and EGF-induced EMT of HNSCC by regulating EGFR/ERK/STAT signaling pathway, indicating its potential future therapeutic application for HNSCC.
Collapse
Affiliation(s)
- Qian Wang
- Oral Disease Research Key Laboratory of Guizhou Tertiary Institution, School of Stomatology, Zunyi Medical University, 563000, Zunyi, Guizhou Province, China
| | - Chengcheng Liao
- Oral Disease Research Key Laboratory of Guizhou Tertiary Institution, School of Stomatology, Zunyi Medical University, 563000, Zunyi, Guizhou Province, China.,Department of Orthodontics II, Affiliated Stomatological Hospital of Zunyi Medical University, 563000, Zunyi, Guizhou Province, China
| | - Zhangxue Tan
- Department of Orthodontics II, Affiliated Stomatological Hospital of Zunyi Medical University, 563000, Zunyi, Guizhou Province, China
| | - Xiaolan Li
- Microbial Resources and Drug Development Key Laboratory of Guizhou Tertiary Institution, Life Sciences Institute, Zunyi Medical University, 563000, Zunyi, China
| | - Xiaoyan Guan
- Department of Orthodontics II, Affiliated Stomatological Hospital of Zunyi Medical University, 563000, Zunyi, Guizhou Province, China
| | - Hao Li
- Department of Orthodontics II, Affiliated Stomatological Hospital of Zunyi Medical University, 563000, Zunyi, Guizhou Province, China
| | - Zhongjia Tian
- Department of Orthodontics II, Affiliated Stomatological Hospital of Zunyi Medical University, 563000, Zunyi, Guizhou Province, China
| | - Jianguo Liu
- Oral Disease Research Key Laboratory of Guizhou Tertiary Institution, School of Stomatology, Zunyi Medical University, 563000, Zunyi, Guizhou Province, China.
| | - Jiaxing An
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, 563000, Zunyi, Guizhou Province, China.
| |
Collapse
|
28
|
Lv G, Zhang L, Gao L, Cui J, Liu Z, Sun B, Wang G, Tang Q. The application of single-cell sequencing in pancreatic neoplasm: analysis, diagnosis and treatment. Br J Cancer 2023; 128:206-218. [PMID: 36307645 PMCID: PMC9902442 DOI: 10.1038/s41416-022-02023-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 10/11/2022] [Accepted: 10/12/2022] [Indexed: 02/08/2023] Open
Abstract
Pancreatic neoplasms, including pancreatic ductal adenocarcinoma (PDAC), intraductal papillary mucinous neoplasm (IPMN) and pancreatic cystic neoplasms (PCNs), are the most puzzling diseases. Numerous studies have not brought significant improvements in prognosis and diagnosis, especially in PDAC. One important reason is that previous studies only focused on differences between patients and healthy individuals but ignored intratumoral heterogeneity. In recent years, single-cell sequencing techniques, represented by single-cell RNA sequencing (scRNA-seq), have emerged by which researchers can analyse each cell in tumours instead of their average levels. Herein, we summarise the new current knowledge of single-cell sequencing in pancreatic neoplasms with respect to techniques, tumour heterogeneities and treatments.
Collapse
Affiliation(s)
- Gaoyuan Lv
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, 150000, Harbin, Heilongjiang Province, China
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Liang Zhang
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, 150000, Harbin, Heilongjiang Province, China
| | - Lei Gao
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, 150000, Harbin, Heilongjiang Province, China
| | - Jitao Cui
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, 150000, Harbin, Heilongjiang Province, China
| | - Ziying Liu
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, 150000, Harbin, Heilongjiang Province, China
| | - Bei Sun
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, 150000, Harbin, Heilongjiang Province, China
| | - Gang Wang
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, 150000, Harbin, Heilongjiang Province, China.
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China.
| | - Qiushi Tang
- Chinese Journal of Practical Surgery, Chinese Medical University, Shenyang, China.
| |
Collapse
|
29
|
Connecting multiple microenvironment proteomes uncovers the biology in head and neck cancer. Nat Commun 2022; 13:6725. [PMID: 36344512 PMCID: PMC9640649 DOI: 10.1038/s41467-022-34407-1] [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: 01/21/2022] [Accepted: 10/25/2022] [Indexed: 11/09/2022] Open
Abstract
The poor prognosis of head and neck cancer (HNC) is associated with metastasis within the lymph nodes (LNs). Herein, the proteome of 140 multisite samples from a 59-HNC patient cohort, including primary and matched LN-negative or -positive tissues, saliva, and blood cells, reveals insights into the biology and potential metastasis biomarkers that may assist in clinical decision-making. Protein profiles are strictly associated with immune modulation across datasets, and this provides the basis for investigating immune markers associated with metastasis. The proteome of LN metastatic cells recapitulates the proteome of the primary tumor sites. Conversely, the LN microenvironment proteome highlights the candidate prognostic markers. By integrating prioritized peptide, protein, and transcript levels with machine learning models, we identify nodal metastasis signatures in blood and saliva. We present a proteomic characterization wiring multiple sites in HNC, thus providing a promising basis for understanding tumoral biology and identifying metastasis-associated signatures.
Collapse
|
30
|
Yang J, Aljitawi O, Van Veldhuizen P. Prostate Cancer Stem Cells: The Role of CD133. Cancers (Basel) 2022; 14:5448. [PMID: 36358865 PMCID: PMC9656005 DOI: 10.3390/cancers14215448] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 10/17/2022] [Accepted: 11/01/2022] [Indexed: 09/27/2023] Open
Abstract
Prostate cancer stem cells (PCSCs), possessing self-renewal properties and resistance to anticancer treatment, are possibly the leading cause of distant metastasis and treatment failure in prostate cancer (PC). CD133 is one of the most well-known and valuable cell surface markers of cancer stem cells (CSCs) in many cancers, including PC. In this article, we focus on reviewing the role of CD133 in PCSC. Any other main stem cell biomarkers in PCSC reported from key publications, as well as about vital research progress of CD133 in CSCs of different cancers, will be selectively reviewed to help us inform the main topic.
Collapse
Affiliation(s)
| | - Omar Aljitawi
- Department of Medicine, Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Peter Van Veldhuizen
- Department of Medicine, Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY 14642, USA
| |
Collapse
|
31
|
Peng J, Li F, Wang J, Wang C, Jiang Y, Liu B, He J, Yuan K, Pan C, Lin M, Zhou B, Chen L, Gao D, Zhao Y. Identification of a rare Gli1 + progenitor cell population contributing to liver regeneration during chronic injury. Cell Discov 2022; 8:118. [PMID: 36316325 PMCID: PMC9622734 DOI: 10.1038/s41421-022-00474-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 09/21/2022] [Indexed: 11/06/2022] Open
Abstract
In adults, hepatocytes are mainly replenished from the existing progenitor pools of hepatocytes and cholangiocytes during chronic liver injury. However, it is unclear whether other cell types in addition to classical hepatocytes and cholangiocytes contribute to hepatocyte regeneration after chronic liver injuries. Here, we identified a new biphenotypic cell population that contributes to hepatocyte regeneration during chronic liver injuries. We found that a cell population expressed Gli1 and EpCAM (EpCAM+Gli1+), which was further characterized with both epithelial and mesenchymal identities by single-cell RNA sequencing. Genetic lineage tracing using dual recombinases revealed that Gli1+ nonhepatocyte cell population could generate hepatocytes after chronic liver injury. EpCAM+Gli1+ cells exhibited a greater capacity for organoid formation with functional hepatocytes in vitro and liver regeneration upon transplantation in vivo. Collectively, these findings demonstrate that EpCAM+Gli1+ cells can serve as a new source of liver progenitor cells and contribute to liver repair and regeneration.
Collapse
Affiliation(s)
- Jiayin Peng
- grid.9227.e0000000119573309State Key Laboratory of Molecular Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
| | - Fei Li
- grid.9227.e0000000119573309State Key Laboratory of Molecular Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
| | - Jia Wang
- grid.9227.e0000000119573309State Key Laboratory of Molecular Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China ,grid.410726.60000 0004 1797 8419University of Chinese Academy of Sciences, Beijing, China
| | - Chaoxiong Wang
- grid.9227.e0000000119573309State Key Laboratory of Molecular Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China ,grid.410726.60000 0004 1797 8419University of Chinese Academy of Sciences, Beijing, China
| | - Yiao Jiang
- grid.9227.e0000000119573309State Key Laboratory of Molecular Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China ,grid.410726.60000 0004 1797 8419University of Chinese Academy of Sciences, Beijing, China
| | - Biao Liu
- grid.9227.e0000000119573309State Key Laboratory of Molecular Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China ,grid.410726.60000 0004 1797 8419University of Chinese Academy of Sciences, Beijing, China
| | - Juan He
- grid.9227.e0000000119573309State Key Laboratory of Molecular Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China ,grid.410726.60000 0004 1797 8419University of Chinese Academy of Sciences, Beijing, China
| | - Kai Yuan
- grid.9227.e0000000119573309State Key Laboratory of Molecular Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
| | - Chenyu Pan
- grid.24516.340000000123704535Department of General Surgery, Yangpu Hospital, Tongji University School of Medicine, Shanghai, China
| | - Moubin Lin
- grid.24516.340000000123704535Department of General Surgery, Yangpu Hospital, Tongji University School of Medicine, Shanghai, China
| | - Bin Zhou
- grid.9227.e0000000119573309State Key Laboratory of Molecular Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
| | - Luonan Chen
- grid.9227.e0000000119573309State Key Laboratory of Molecular Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
| | - Dong Gao
- grid.9227.e0000000119573309State Key Laboratory of Molecular Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China ,grid.9227.e0000000119573309Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
| | - Yun Zhao
- grid.9227.e0000000119573309State Key Laboratory of Molecular Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China ,grid.440637.20000 0004 4657 8879School of Life Science and Technology, ShanghaiTech University, Shanghai, China ,grid.410726.60000 0004 1797 8419Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, Zhejiang China
| |
Collapse
|
32
|
Lee J, Pang K, Kim J, Hong E, Lee J, Cho HJ, Park J, Son M, Park S, Lee M, Ooshima A, Park KS, Yang HK, Yang KM, Kim SJ. ESRP1-regulated isoform switching of LRRFIP2 determines metastasis of gastric cancer. Nat Commun 2022; 13:6274. [PMID: 36307405 PMCID: PMC9616898 DOI: 10.1038/s41467-022-33786-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 10/03/2022] [Indexed: 12/25/2022] Open
Abstract
Although accumulating evidence indicates that alternative splicing is aberrantly altered in many cancers, the functional mechanism remains to be elucidated. Here, we show that epithelial and mesenchymal isoform switches of leucine-rich repeat Fli-I-interacting protein 2 (LRRFIP2) regulated by epithelial splicing regulatory protein 1 (ESRP1) correlate with metastatic potential of gastric cancer cells. We found that expression of the splicing variants of LRRFIP2 was closely correlated with that of ESRP1. Surprisingly, ectopic expression of the mesenchymal isoform of LRRFIP2 (variant 3) dramatically increased liver metastasis of gastric cancer cells, whereas deletion of exon 7 of LRRFIP2 by the CRISPR/Cas9 system caused an isoform switch, leading to marked suppression of liver metastasis. Mechanistically, the epithelial LRRFIP2 isoform (variant 2) inhibited the oncogenic function of coactivator-associated arginine methyltransferase 1 (CARM1) through interaction. Taken together, our data reveals a mechanism of LRRFIP2 isoform switches in gastric cancer with important implication for cancer metastasis.
Collapse
Affiliation(s)
- Jihee Lee
- GILO Institute, GILO Foundation, Seoul, 06668 Korea ,grid.410886.30000 0004 0647 3511Department of Biomedical Science, College of Life Science, CHA University, Seongnam, Gyeonggi-do 13488 Korea
| | | | - Junil Kim
- grid.263765.30000 0004 0533 3568School of Systems Biomedical Science, Soongsil University, Seoul, 06978 Korea
| | - Eunji Hong
- GILO Institute, GILO Foundation, Seoul, 06668 Korea ,grid.264381.a0000 0001 2181 989XDepartment of Biomedical Science, College of Life Science, Sungkyunkwan University, Suwon, Gyeonggi-do 16419 Korea
| | - Jeeyun Lee
- grid.264381.a0000 0001 2181 989XDivision of Hematology-Oncology, Department of Medicine, Samsung Medical Center Sungkyunkwan University School of Medicine, Seoul, 06351 Korea
| | - Hee Jin Cho
- grid.258803.40000 0001 0661 1556Department of Biomedical Convergence Science and Technology, Kyungpook National University, Daegu, 41566 Korea ,grid.414964.a0000 0001 0640 5613Innovative Therapeutic Research Center, Precision Medicine Research Institute, Samsung Medical Center, Seoul, 06531 Republic of Korea
| | - Jinah Park
- GILO Institute, GILO Foundation, Seoul, 06668 Korea
| | - Minjung Son
- GILO Institute, GILO Foundation, Seoul, 06668 Korea ,grid.264381.a0000 0001 2181 989XDepartment of Biomedical Science, College of Life Science, Sungkyunkwan University, Suwon, Gyeonggi-do 16419 Korea
| | - Sihyun Park
- GILO Institute, GILO Foundation, Seoul, 06668 Korea
| | | | | | - Kyung-Soon Park
- grid.410886.30000 0004 0647 3511Department of Biomedical Science, College of Life Science, CHA University, Seongnam, Gyeonggi-do 13488 Korea
| | - Han-Kwang Yang
- grid.412484.f0000 0001 0302 820XDepartment of Surgery, Seoul National University Hospital, Seoul, 03080 Korea ,grid.31501.360000 0004 0470 5905Cancer Research Institute, Seoul National University, Seoul, 03080 Korea
| | | | - Seong-Jin Kim
- GILO Institute, GILO Foundation, Seoul, 06668 Korea ,Medpacto Inc., Seoul, 06668 Korea
| |
Collapse
|
33
|
Choi Y, Park NJY, Le TM, Lee E, Lee D, Nguyen HDT, Cho J, Park JY, Han HS, Chong GO. Immune Pathway and Gene Database (IMPAGT) Revealed the Immune Dysregulation Dynamics and Overactivation of the PI3K/Akt Pathway in Tumor Buddings of Cervical Cancer. Curr Issues Mol Biol 2022; 44:5139-5152. [PMID: 36354662 PMCID: PMC9688570 DOI: 10.3390/cimb44110350] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 10/17/2022] [Accepted: 10/21/2022] [Indexed: 08/31/2023] Open
Abstract
Tumor budding (TB) is a small cluster of malignant cells at the invasive front of a tumor. Despite being an adverse prognosis marker, little research has been conducted on the tumor immune microenvironment of tumor buddings, especially in cervical cancer. Therefore, RNA sequencing was performed using 21 formalin-fixed, paraffin-embedded slides of cervical tissues, and differentially expressed genes (DEGs) were analyzed. Immune Pathway and Gene Database (IMPAGT) was generated for immune profiling. "Pathway in Cancer" was identified as the most enriched pathway for both up- and downregulated DEGs. Kyoto Encyclopedia of Genes and Genomes Mapper and Gene Ontology further revealed the activation of the PI3K/Akt signaling pathway. An IMPAGT analysis revealed immune dysregulation even at the tumor budding stage, especially in the PI3K/Akt/mTOR axis, with a high efficiency and integrity. These findings emphasized the clinical significance of tumor buddings and the necessity of blocking the overactivation of the PI3K/Akt/mTOR pathway to improve targeted therapy in cervical cancer.
Collapse
Affiliation(s)
- Yeseul Choi
- Department of Biomedical Science, Graduate School, Kyungpook National University, Daegu 41944, Korea
- BK21 Four Program, School of Medicine, Kyungpook National University, Daegu 41944, Korea
| | - Nora Jee-Young Park
- Department of Pathology, School of Medicine, Kyungpook National University, Daegu 41944, Korea
- Department of Pathology, Kyungpook National University Chilgok Hospital, Daegu 41404, Korea
- Clinical Omics Institute, Kyungpook National University, Daegu 41405, Korea
| | - Tan Minh Le
- Department of Biomedical Science, Graduate School, Kyungpook National University, Daegu 41944, Korea
- BK21 Four Program, School of Medicine, Kyungpook National University, Daegu 41944, Korea
| | - Eunmi Lee
- Department of Biomedical Science, Graduate School, Kyungpook National University, Daegu 41944, Korea
- BK21 Four Program, School of Medicine, Kyungpook National University, Daegu 41944, Korea
| | - Donghyeon Lee
- Department of Biomedical Science, Graduate School, Kyungpook National University, Daegu 41944, Korea
- BK21 Four Program, School of Medicine, Kyungpook National University, Daegu 41944, Korea
| | - Hong Duc Thi Nguyen
- Department of Biomedical Science, Graduate School, Kyungpook National University, Daegu 41944, Korea
- BK21 Four Program, School of Medicine, Kyungpook National University, Daegu 41944, Korea
| | - Junghwan Cho
- Clinical Omics Institute, Kyungpook National University, Daegu 41405, Korea
| | - Ji-Young Park
- Department of Pathology, School of Medicine, Kyungpook National University, Daegu 41944, Korea
- Department of Pathology, Kyungpook National University Chilgok Hospital, Daegu 41404, Korea
| | - Hyung Soo Han
- Department of Biomedical Science, Graduate School, Kyungpook National University, Daegu 41944, Korea
- BK21 Four Program, School of Medicine, Kyungpook National University, Daegu 41944, Korea
- Clinical Omics Institute, Kyungpook National University, Daegu 41405, Korea
- Department of Physiology, School of Medicine, Kyungpook National University, Daegu 41944, Korea
| | - Gun Oh Chong
- Clinical Omics Institute, Kyungpook National University, Daegu 41405, Korea
- Department of Obstetrics and Gynecology, School of Medicine, Kyungpook National University, Daegu 41944, Korea
- Department of Obstetrics and Gynecology, Kyungpook National University Chilgok Hospital, Daegu 41404, Korea
| |
Collapse
|
34
|
Xiao RR, Jin L, Xie N, Luo P, Gao W, Tu P, Ai X. Establishment and large-scale validation of a three-dimensional tumor model on an array chip for anticancer drug evaluation. Front Pharmacol 2022; 13:1032975. [PMID: 36313330 PMCID: PMC9596801 DOI: 10.3389/fphar.2022.1032975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 09/28/2022] [Indexed: 11/29/2022] Open
Abstract
Two-dimensional (2D) tumor model has always poorly predicted drug response of animal model due to the lack of recapitulation of tumor microenvironment. Establishing a biomimetic, controllable, and cost-effective three-dimensional (3D) model and large-scale validation of its in vivo predictivity has shown promise in bridging the gap between the 2D tumor model and animal model. Here, we established a matrigel-based 3D micro-tumor model on an array chip for large-scale anticancer drug evaluation. Compared with the 2D tumor model, the 3D tumor model on the chip showed spheroid morphology, slower proliferation kinetics, and comparable reproducibility. Next, the results of the chemotherapeutic evaluation from 18 drugs against 27 cancer cell lines showed 17.6% of drug resistance on the 3D tumor model. Moreover, the evaluation results of targeted drugs showed expected sensitivity and higher specificity on the 3D tumor model compared with the 2D model. Finally, the evaluation results on the 3D tumor model were more consistent with the in vivo cell-derived xenograft model, and excluded 95% false-positive results from the 2D model. Overall, the matrigel-based 3D micro-tumor model on the array chip provides a promising tool to accelerate anticancer drug discovery.
Collapse
Affiliation(s)
- Rong-Rong Xiao
- R&D Department, Beijing Daxiang Biotech Co., Ltd., Beijing, China
| | - Lei Jin
- Oncology and Immunology Unit, WuXi Biology, WuXi AppTec (Shanghai) Co., Ltd., Shanghai, China
| | - Nan Xie
- Oncology and Immunology Unit, WuXi Biology, WuXi AppTec (Shanghai) Co., Ltd., Shanghai, China
| | - Piaopiao Luo
- R&D Department, Beijing Daxiang Biotech Co., Ltd., Beijing, China
| | - Wenjie Gao
- Oncology and Immunology Unit, WuXi Biology, WuXi AppTec (Shanghai) Co., Ltd., Shanghai, China
| | - Pengfei Tu
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Xiaoni Ai
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
- *Correspondence: Xiaoni Ai,
| |
Collapse
|
35
|
Silva ATF, Rodrigues CM, Ferreira ICC, Santos LLD, Santos DW, Araújo TG, Canto PPL, Paiva CE, Goulart LR, Maia YCP. A Novel Detection Method of Breast Cancer through a Simple Panel of Biomarkers. Int J Mol Sci 2022; 23:ijms231911983. [PMID: 36233281 PMCID: PMC9570447 DOI: 10.3390/ijms231911983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 09/25/2022] [Accepted: 09/28/2022] [Indexed: 11/23/2022] Open
Abstract
Circulating tumor cells (CTCs) have been identified as responsible for the spread of tumors to other organs of the body. In this sense, the development of sensitive and specific assays for their detection is important to reduce the number of deaths due to metastases. Here, we assessed whether the detection of CTCs in peripheral blood can serve in the construction of a panel of diagnosis and monitoring treatments of breast cancer (BC), focusing on the expression of markers of epithelial-mesenchymal transition. Through analyzing the blood from women without breast alterations (control), women with benign alterations, women with breast cancer without chemotherapy, and women with breast cancer with chemotherapy, we identified the best markers by transcriptional levels and determined three profiles of CTCs (mesenchymal, intermediate, and epithelial) by flow cytometry which, combined, can be used for diagnosis and therapy monitoring with sensitivity and specificity between 80% and 100%. Therefore, we have developed a method for detecting breast cancer based on the analysis of CTC profiles by epithelial-mesenchymal transition markers which, combined, can be used for the diagnosis and monitoring of therapy.
Collapse
Affiliation(s)
- Alinne T. F. Silva
- Molecular Biology and Nutrition Research Group, School of Medicine, Graduate Program in Health Science, Av. Amazonas sn, Block 2E, 2º Floor, Room 210, Campus Umuarama, Uberlandia 38405-320, Minas Gerais, Brazil
- Laboratory of Nanobiotechnology Prof. Dr. Luiz Ricardo Goulart Filho, Institute of Biotechnology, Federal University of Uberlandia, Av. Amazonas sn, Block 2E, 2º Floor, Room 248, Campus Umuarama, Uberlandia 38405-302, Minas Gerais, Brazil
| | - Cláudia M. Rodrigues
- Laboratory of Nanobiotechnology Prof. Dr. Luiz Ricardo Goulart Filho, Institute of Biotechnology, Federal University of Uberlandia, Av. Amazonas sn, Block 2E, 2º Floor, Room 248, Campus Umuarama, Uberlandia 38405-302, Minas Gerais, Brazil
| | - Izabella C. C. Ferreira
- Molecular Biology and Nutrition Research Group, School of Medicine, Graduate Program in Health Science, Av. Amazonas sn, Block 2E, 2º Floor, Room 210, Campus Umuarama, Uberlandia 38405-320, Minas Gerais, Brazil
- Laboratory of Nanobiotechnology Prof. Dr. Luiz Ricardo Goulart Filho, Institute of Biotechnology, Federal University of Uberlandia, Av. Amazonas sn, Block 2E, 2º Floor, Room 248, Campus Umuarama, Uberlandia 38405-302, Minas Gerais, Brazil
| | - Letícia L. D. Santos
- Molecular Biology and Nutrition Research Group, School of Medicine, Graduate Program in Health Science, Av. Amazonas sn, Block 2E, 2º Floor, Room 210, Campus Umuarama, Uberlandia 38405-320, Minas Gerais, Brazil
- Laboratory of Nanobiotechnology Prof. Dr. Luiz Ricardo Goulart Filho, Institute of Biotechnology, Federal University of Uberlandia, Av. Amazonas sn, Block 2E, 2º Floor, Room 248, Campus Umuarama, Uberlandia 38405-302, Minas Gerais, Brazil
| | - Donizeti W. Santos
- Obstetric Division, University Hospital, Federal University of Uberlandia, Av. Pará, 1720, Block 2H, Campus Umuarama, Uberlandia 38405-320, Minas Gerais, Brazil
| | - Thaise G. Araújo
- Laboratory of Nanobiotechnology Prof. Dr. Luiz Ricardo Goulart Filho, Institute of Biotechnology, Federal University of Uberlandia, Av. Amazonas sn, Block 2E, 2º Floor, Room 248, Campus Umuarama, Uberlandia 38405-302, Minas Gerais, Brazil
| | - Paula P. L. Canto
- Department of Clinical Oncology, Clinics Hospital, Federal University of Uberlandia, Av. Pará, 1720, Oncology Sector, Room 9, Campus Umuarama, Uberlandia 38405-320, Minas Gerais, Brazil
| | - Carlos E. Paiva
- Department of Clinical Oncology, Graduate Program in Oncology, Palliative Care and Quality of Life Research Group (GPQual), Barretos Cancer Hospital, R. Antenor Duarte Vilela, 1331, Doutor Paulo Prata, Barretos 14784-400, Sao Paulo, Brazil
| | - Luiz R. Goulart
- Molecular Biology and Nutrition Research Group, School of Medicine, Graduate Program in Health Science, Av. Amazonas sn, Block 2E, 2º Floor, Room 210, Campus Umuarama, Uberlandia 38405-320, Minas Gerais, Brazil
- Laboratory of Nanobiotechnology Prof. Dr. Luiz Ricardo Goulart Filho, Institute of Biotechnology, Federal University of Uberlandia, Av. Amazonas sn, Block 2E, 2º Floor, Room 248, Campus Umuarama, Uberlandia 38405-302, Minas Gerais, Brazil
| | - Yara C. P. Maia
- Molecular Biology and Nutrition Research Group, School of Medicine, Graduate Program in Health Science, Av. Amazonas sn, Block 2E, 2º Floor, Room 210, Campus Umuarama, Uberlandia 38405-320, Minas Gerais, Brazil
- Laboratory of Nanobiotechnology Prof. Dr. Luiz Ricardo Goulart Filho, Institute of Biotechnology, Federal University of Uberlandia, Av. Amazonas sn, Block 2E, 2º Floor, Room 248, Campus Umuarama, Uberlandia 38405-302, Minas Gerais, Brazil
- Correspondence: ; Tel.: +34-3225-8628
| |
Collapse
|
36
|
Interplay between Partial EMT and Cisplatin Resistance as the Drivers for Recurrence in HNSCC. Biomedicines 2022; 10:biomedicines10102482. [PMID: 36289744 PMCID: PMC9598677 DOI: 10.3390/biomedicines10102482] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 09/26/2022] [Accepted: 09/27/2022] [Indexed: 11/20/2022] Open
Abstract
This study aims to investigate the role of partial epithelial to mesenchymal transition (pEMT)-related proteins in modulating Cisplatin resistance in head and neck squamous cell carcinoma (HNSCC). SCC-25 cells were pre-treated with TGF-beta1 followed by transient Krüppel-like Factor 4 (KLF4)-overexpression and Cisplatin treatment. Cell growth, cell morphological changes and cell migration were assessed using Juli BR live cell video-microscopy. In addition, Ki-67 and Slug immunostaining and follow-up image cytometric analysis of primary and recurrent HNSCC tumors were performed to evaluate the proliferation index (PI) and the EMT-like phenotype. We observed that proliferating and Slug-positive tumor cells expand after therapy in HNSCC. Subsequently, protein analysis revealed the stabilization of Slug, upregulation of Vimentin and phospho-p38 (p-p38) in Cisplatin-resistant SCC-25 cells. Moreover, KLF4-overexpression contributed to Cisplatin sensitivity by reduction of Slug at the protein level. This work strongly suggests that an pEMT-like pathway is activated in recurrent and Cisplatin-resistant HNSCC. Finally, stable KLF4-overexpression might sensitize HNSCC tumor cells for Cisplatin treatment.
Collapse
|
37
|
Marles H, Biddle A. Cancer stem cell plasticity and its implications in the development of new clinical approaches for oral squamous cell carcinoma. Biochem Pharmacol 2022; 204:115212. [PMID: 35985402 DOI: 10.1016/j.bcp.2022.115212] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 08/10/2022] [Accepted: 08/10/2022] [Indexed: 11/25/2022]
Abstract
Oral squamous cell carcinoma (SCC) represents a major worldwide disease burden, with high rates of recurrence and metastatic spread following existing treatment methods. Populations of treatment resistant cancer stem cells (CSCs) are well characterised in oral SCC. These populations of CSCs engage the cellular programme known as epithelial mesenchymal transition (EMT) to enhance metastatic spread and therapeutic resistance. EMT is characterised by specific morphological changes and the expression of certain cell surface markers that represent a transition from an epithelial phenotype to a mesenchymal phenotype. This process is regulated by several cellular pathways that interact both horizontally and hierarchically. The cellular changes in EMT occur along a spectrum, with sub-populations of cells displaying both epithelial and mesenchymal features. The unique features of these CSCs in terms of their EMT state, cell surface markers and metabolism may offer new druggable targets. In addition, these features could be used to identify more aggressive disease states and the opportunity to personalise therapy depending on the presence of certain CSC sub-populations.
Collapse
Affiliation(s)
- Henry Marles
- Centre for Cell Biology and Cutaneous Research, Blizard Institute, Queen Mary University of London, London E1 2AT, UK
| | - Adrian Biddle
- Centre for Cell Biology and Cutaneous Research, Blizard Institute, Queen Mary University of London, London E1 2AT, UK.
| |
Collapse
|
38
|
Pelosi G, De Luca M, Cannone M, Balladore E, Ricotti I, Toniolo D, Incarbone M. Metastatic mucinous ovarian carcinoma simulating lung primary: an integrated diagnostic lesson. Pathologica 2022; 114:365-372. [PMID: 36305022 PMCID: PMC9614303 DOI: 10.32074/1591-951x-802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Accepted: 09/19/2022] [Indexed: 11/10/2022] Open
Abstract
We herein document a rare instance of primary mucinous ovarian carcinoma metastatic to the left lung, whose deceptive secondary derivation was already envisaged according to the spectacular thromboembolism involving small pulmonary vessels, thereby realizing a centrifugal and centripetal metastatizing loop. This presentation was indicative of dismal prognosis. A multimodal biomarker key approach is herein emphasized, which included close clinico-pathologic data integration.
Collapse
Affiliation(s)
- Giuseppe Pelosi
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy,Inter-hospital Pathology Division, Istituto di Ricovero e Cura a Carattere Scentifico - IRCCS MultiMedica, Milan, Italy,Correspondence Giuseppe Pelosi Inter-hospital Pathology Division, Science & Technology Park, IRCCS MultiMedica, via G. Fantoli 16/15, 20138 Milan, Italy Tel.: + 39 02 5540 6572/6509 Fax: +39 02 5540 6570 E-mail:
| | - Marco De Luca
- Inter-hospital Pathology Division, Istituto di Ricovero e Cura a Carattere Scentifico - IRCCS MultiMedica, Milan, Italy
| | - Maria Cannone
- Inter-hospital Pathology Division, Istituto di Ricovero e Cura a Carattere Scentifico - IRCCS MultiMedica, Milan, Italy
| | - Emanuela Balladore
- Inter-hospital Pathology Division, Istituto di Ricovero e Cura a Carattere Scentifico - IRCCS MultiMedica, Milan, Italy
| | - Isabella Ricotti
- Division of Pathologic Anatomy and Histology, Azienda Socio-Sanitaria Territoriale Rhodense - Presidio Ospedaliero di Rho, Rho, Italy
| | - Davide Toniolo
- Division of Medical Oncology and Hematology, Azienda Socio-Sanitaria Territoriale Rhodense - Presidio Ospedaliero di Rho, Rho, Italy
| | - Matteo Incarbone
- Division of Thoracic Surgery, Istituto di Ricovero e Cura a Carattere Scentifico - IRCCS MultiMedica, Milan, Italy
| |
Collapse
|
39
|
Glycosphingolipids are mediators of cancer plasticity through independent signaling pathways. Cell Rep 2022; 40:111181. [PMID: 35977490 DOI: 10.1016/j.celrep.2022.111181] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 06/01/2022] [Accepted: 07/19/2022] [Indexed: 11/23/2022] Open
Abstract
The molecular repertoire promoting cancer cell plasticity is not fully elucidated. Here, we propose that glycosphingolipids (GSLs), specifically the globo and ganglio series, correlate and promote the transition between epithelial and mesenchymal cells. The epithelial character of ovarian cancer remains stable throughout disease progression, and spatial glycosphingolipidomics reveals elevated globosides in the tumor compartment compared with the ganglioside-rich stroma. CRISPR-Cas9 knockin mediated truncation of endogenous E-cadherin induces epithelial-to-mesenchymal transition (EMT) and decreases globosides. The transcriptomics analysis identifies the ganglioside-synthesizing enzyme ST8SIA1 to be consistently elevated in mesenchymal-like samples, predicting poor outcome. Subsequent deletion of ST8SIA1 induces epithelial cell features through mTORS2448 phosphorylation, whereas loss of globosides in ΔA4GALT cells, resulting in EMT, is accompanied by increased ERKY202/T204 and AKTS124. The GSL composition dynamics corroborate cancer cell plasticity, and further evidence suggests that mesenchymal cells are maintained through ganglioside-dependent, calcium-mediated mechanisms.
Collapse
|
40
|
FMR1NB Involved in Glioma Tumorigenesis Is a Promising Target for Prognosis and Therapy. Curr Med Sci 2022; 42:803-816. [PMID: 35819657 DOI: 10.1007/s11596-022-2586-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 11/12/2021] [Indexed: 11/03/2022]
|
41
|
Xu C, He XY, Ren XH, Han D, Cheng SX. Detection of mRNAs of Ribosomal Protein L15 and E-Cadherin in Living Circulating Tumor Cells at Single Cell Resolution To Study Tumor Heterogeneity. Anal Chem 2022; 94:10610-10616. [PMID: 35856393 DOI: 10.1021/acs.analchem.2c00488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
To study the heterogeneity of circulating tumor cells (CTCs) is of crucial importance to analyze cancer progression and metastasis. However, in situ detection of highly heterogeneous CTCs in peripheral blood still faces an elusive challenge. Here, we show direct detection of two metastasis-related mRNAs of diverse CTCs in whole blood by a triple-targeting nanoprobe. In the nanoprobe, two kinds of molecular beacons, MB1 to detect RPL15 mRNA and MB2 to detect E-cadherin (E-cad) mRNA, are loaded in a highly efficient delivery vector decorated with EpCAM-targeted SYL3C, EGFR-targeted CL4, and CD44-targeted hyaluronic acid chains to specifically deliver MB1/MB2 into epithelial, mesenchymal, and stem CTCs in unprocessed peripheral blood. The numbers of RPL15+ and E-cad+ CTCs are positively correlated with the metastasis stages of cancer patients. This study provides an effective strategy to realize direct observation on diverse metastasis-related genes in living CTCs with different phenotypes to provide accurate information on cancer heterogeneity and metastasis.
Collapse
Affiliation(s)
- Chang Xu
- Key Laboratory of Biomedical Polymers of Ministry of Education, Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China
| | - Xiao-Yan He
- School of Life Sciences, Anhui Medical University, Hefei 230032, P. R. China
| | - Xiao-He Ren
- Key Laboratory of Biomedical Polymers of Ministry of Education, Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China
| | - Di Han
- Key Laboratory of Biomedical Polymers of Ministry of Education, Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China
| | - Si-Xue Cheng
- Key Laboratory of Biomedical Polymers of Ministry of Education, Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China
| |
Collapse
|
42
|
Revisiting Epithelial Carcinogenesis. Int J Mol Sci 2022; 23:ijms23137437. [PMID: 35806442 PMCID: PMC9267463 DOI: 10.3390/ijms23137437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 06/22/2022] [Accepted: 06/22/2022] [Indexed: 12/04/2022] Open
Abstract
The origin of cancer remains one of the most important enigmas in modern biology. This paper presents a hypothesis for the origin of carcinomas in which cellular aging and inflammation enable the recovery of cellular plasticity, which may ultimately result in cancer. The hypothesis describes carcinogenesis as the result of the dedifferentiation undergone by epithelial cells in hyperplasia due to replicative senescence towards a mesenchymal cell state with potentially cancerous behavior. In support of this hypothesis, the molecular, cellular, and histopathological evidence was critically reviewed and reinterpreted when necessary to postulate a plausible generic series of mechanisms for the origin and progression of carcinomas. In addition, the implications of this theoretical framework for the current strategies of cancer treatment are discussed considering recent evidence of the molecular events underlying the epigenetic switches involved in the resistance of breast carcinomas. The hypothesis also proposes an epigenetic landscape for their progression and a potential mechanism for restraining the degree of dedifferentiation and malignant behavior. In addition, the manuscript revisits the gradual degeneration of the nonalcoholic fatty liver disease to propose an integrative generalized mechanistic explanation for the involution and carcinogenesis of tissues associated with aging. The presented hypothesis might serve to understand and structure new findings into a more encompassing view of the genesis of degenerative diseases and may inspire novel approaches for their study and therapy.
Collapse
|
43
|
Shen C, Han L, Liu B, Zhang G, Cai Z, Yin X, Yin Y, Chen Z, Zhang B. The KDM6A-SPARCL1 axis blocks metastasis and regulates the tumour microenvironment of gastrointestinal stromal tumours by inhibiting the nuclear translocation of p65. Br J Cancer 2022; 126:1457-1469. [PMID: 35136209 PMCID: PMC9090789 DOI: 10.1038/s41416-022-01728-3] [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: 06/22/2021] [Revised: 01/07/2022] [Accepted: 01/28/2022] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND It is urgent to explore the pathogenic mechanism of gastrointestinal stromal tumours (GISTs). KDM6A, a histone demethylase, can activate gene transcription and has not been reported in GISTs. SPARCL1 may serve as a metastasis marker in GIST, but the molecular mechanism remains to be further explored. This study aimed to explore the biological function and molecular mechanism of KDM6A and SPARCL1 in GIST. METHODS CCK-8, live cell count, colony formation, wound-healing and Transwell migration and invasion assays were employed to detect the cell proliferation, migration and invasion. A xenograft model and hepatic metastasis model were used to assess the role of KDM6A and SPARCL1 in vivo. RESULTS KDM6A inhibited the proliferation, migration and invasion of GIST cells. Mechanistically, KDM6A promotes the transcription of SPARCL1 by demethylating histone H3 lysine trimethylation and consequently leads to the inactivation of p65. SPARCL1 affected the metastasis of GIST cells in a mesenchymal-epithelial transition- and matrix-metalloproteinase-dependent manner. SPARCL1 knockdown promoted angiogenesis, M2 polarisation and macrophage recruitment by inhibiting the phosphorylation of p65. Moreover, KDM6A and SPARCL1 inhibited hepatic metastasis and macrophage infiltration in vivo. CONCLUSIONS Our findings establish the critical role of the KDM6A-SPARCL1-p65 axis in restraining the malignancy of GIST.
Collapse
Affiliation(s)
- Chaoyong Shen
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, 610041, Chengdu, Sichuan, China
| | - Luyin Han
- Intensive care unit, West China Hospital, Sichuan University, 610041, Chengdu, Sichuan, China
| | - Baike Liu
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, 610041, Chengdu, Sichuan, China
| | - Guixiang Zhang
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, 610041, Chengdu, Sichuan, China
| | - Zhaolun Cai
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, 610041, Chengdu, Sichuan, China
| | - Xiaonan Yin
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, 610041, Chengdu, Sichuan, China
| | - Yuan Yin
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, 610041, Chengdu, Sichuan, China
| | - Zhixin Chen
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, 610041, Chengdu, Sichuan, China
| | - Bo Zhang
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, 610041, Chengdu, Sichuan, China.
| |
Collapse
|
44
|
Ungefroren H, Thürling I, Färber B, Kowalke T, Fischer T, De Assis LVM, Braun R, Castven D, Oster H, Konukiewitz B, Wellner UF, Lehnert H, Marquardt JU. The Quasimesenchymal Pancreatic Ductal Epithelial Cell Line PANC-1-A Useful Model to Study Clonal Heterogeneity and EMT Subtype Shifting. Cancers (Basel) 2022; 14:cancers14092057. [PMID: 35565186 PMCID: PMC9101310 DOI: 10.3390/cancers14092057] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 04/13/2022] [Accepted: 04/18/2022] [Indexed: 12/18/2022] Open
Abstract
Simple Summary Malignant tumors often escape therapy due to clonal propagation of a subfraction of drug-resistant cancer cells. The underlying phenomenon of intratumoral heterogeneity is driven by epithelial–mesenchymal plasticity (EMP) involving the developmental programs, epithelial–mesenchymal transition (EMT), in which epithelial cells are converted to invasive mesenchymal cells, and the reverse process, mesenchymal–epithelial transition (MET), which allows for metastatic outgrowth at distant sites. For therapeutic targeting of EMP, a better understanding of this process is required; however, cellular models with which to study EMP in pancreatic ductal adenocarcinoma (PDAC) are scarce. Using single-cell clonal analysis, we have found the PDAC cell line, PANC-1, to consist of cells with different E/M phenotypes and functional attributes. Parental PANC-1 cultures could be induced in vitro to shift towards either a more mesenchymal or a more epithelial phenotype, and this bidirectional shift was controlled by the small GTPases RAC1 and RAC1b, together identifying PANC-1 cells as a useful model with which to study EMP. Abstract Intratumoral heterogeneity (ITH) is an intrinsic feature of malignant tumors that eventually allows a subfraction of resistant cancer cells to clonally evolve and cause therapy failure or relapse. ITH, cellular plasticity and tumor progression are driven by epithelial–mesenchymal transition (EMT) and the reverse process, MET. During these developmental programs, epithelial (E) cells are successively converted to invasive mesenchymal (M) cells, or back to E cells, by passing through a series of intermediate E/M states, a phenomenon termed E–M plasticity (EMP). The induction of MET has clinical potential as it can block the initial EMT stages that favor tumor cell dissemination, while its inhibition can curb metastatic outgrowth at distant sites. In pancreatic ductal adenocarcinoma (PDAC), cellular models with which to study EMP or MET induction are scarce. Here, we have generated single cell-derived clonal cultures of the quasimesenchymal PDAC-derived cell line, PANC-1, and found that these differ strongly with respect to cell morphology and EMT marker expression, allowing for their tentative classification as E, E/M or M. Interestingly, the different EMT phenotypes were found to segregate with differences in tumorigenic potential in vitro, as measured by colony forming and invasive activities, and in circadian clock function. Moreover, the individual clones the phenotypes of which remained stable upon prolonged culture also responded differently to treatment with transforming growth factor (TGF)β1 in regard to regulation of growth and individual TGFβ target genes, and to culture conditions that favour ductal-to-endocrine transdifferentiation as a more direct measure for cellular plasticity. Of note, stimulation with TGFβ1 induced a shift in parental PANC-1 cultures towards a more extreme M and invasive phenotype, while exposing the cells to a combination of the proinflammatory cytokines IFNγ, IL1β and TNFα (IIT) elicited a shift towards a more E and less invasive phenotype resembling a MET-like process. Finally, we show that the actions of TGFβ1 and IIT both converge on regulating the ratio of the small GTPase RAC1 and its splice isoform, RAC1b. Our data provide strong evidence for dynamic EMT–MET transitions and qualify this cell line as a useful model with which to study EMP.
Collapse
Affiliation(s)
- Hendrik Ungefroren
- First Department of Medicine, University Hospital Schleswig-Holstein, Campus Lübeck, Center of Brain, Behavior and Metabolism (CBBM), University of Lübeck, D-23538 Lübeck, Germany; (I.T.); (T.K.); (T.F.); (D.C.); (J.-U.M.)
- Clinic for Surgery, University Hospital Schleswig-Holstein, Campus Lübeck, University of Lübeck, D-23538 Lübeck, Germany; (B.F.); (R.B.); (U.F.W.)
- Institute of Pathology, University Hospital Schleswig-Holstein, Campus Kiel, D-24105 Kiel, Germany;
- Correspondence:
| | - Isabel Thürling
- First Department of Medicine, University Hospital Schleswig-Holstein, Campus Lübeck, Center of Brain, Behavior and Metabolism (CBBM), University of Lübeck, D-23538 Lübeck, Germany; (I.T.); (T.K.); (T.F.); (D.C.); (J.-U.M.)
| | - Benedikt Färber
- Clinic for Surgery, University Hospital Schleswig-Holstein, Campus Lübeck, University of Lübeck, D-23538 Lübeck, Germany; (B.F.); (R.B.); (U.F.W.)
| | - Tanja Kowalke
- First Department of Medicine, University Hospital Schleswig-Holstein, Campus Lübeck, Center of Brain, Behavior and Metabolism (CBBM), University of Lübeck, D-23538 Lübeck, Germany; (I.T.); (T.K.); (T.F.); (D.C.); (J.-U.M.)
| | - Tanja Fischer
- First Department of Medicine, University Hospital Schleswig-Holstein, Campus Lübeck, Center of Brain, Behavior and Metabolism (CBBM), University of Lübeck, D-23538 Lübeck, Germany; (I.T.); (T.K.); (T.F.); (D.C.); (J.-U.M.)
| | - Leonardo Vinícius Monteiro De Assis
- Institute for Neurobiology, Center of Brain, Behavior and Metabolism (CBBM), University of Lübeck, D-23538 Lübeck, Germany; (L.V.M.D.A.); (H.O.)
| | - Rüdiger Braun
- Clinic for Surgery, University Hospital Schleswig-Holstein, Campus Lübeck, University of Lübeck, D-23538 Lübeck, Germany; (B.F.); (R.B.); (U.F.W.)
| | - Darko Castven
- First Department of Medicine, University Hospital Schleswig-Holstein, Campus Lübeck, Center of Brain, Behavior and Metabolism (CBBM), University of Lübeck, D-23538 Lübeck, Germany; (I.T.); (T.K.); (T.F.); (D.C.); (J.-U.M.)
| | - Henrik Oster
- Institute for Neurobiology, Center of Brain, Behavior and Metabolism (CBBM), University of Lübeck, D-23538 Lübeck, Germany; (L.V.M.D.A.); (H.O.)
| | - Björn Konukiewitz
- Institute of Pathology, University Hospital Schleswig-Holstein, Campus Kiel, D-24105 Kiel, Germany;
| | - Ulrich Friedrich Wellner
- Clinic for Surgery, University Hospital Schleswig-Holstein, Campus Lübeck, University of Lübeck, D-23538 Lübeck, Germany; (B.F.); (R.B.); (U.F.W.)
| | | | - Jens-Uwe Marquardt
- First Department of Medicine, University Hospital Schleswig-Holstein, Campus Lübeck, Center of Brain, Behavior and Metabolism (CBBM), University of Lübeck, D-23538 Lübeck, Germany; (I.T.); (T.K.); (T.F.); (D.C.); (J.-U.M.)
| |
Collapse
|
45
|
Hoti G, Matencio A, Rubin Pedrazzo A, Cecone C, Appleton SL, Khazaei Monfared Y, Caldera F, Trotta F. Nutraceutical Concepts and Dextrin-Based Delivery Systems. Int J Mol Sci 2022; 23:4102. [PMID: 35456919 PMCID: PMC9031143 DOI: 10.3390/ijms23084102] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 03/26/2022] [Accepted: 04/02/2022] [Indexed: 12/12/2022] Open
Abstract
Nutraceuticals are bioactive or chemical compounds acclaimed for their valuable biological activities and health-promoting effects. The global community is faced with many health concerns such as cancers, cardiovascular and neurodegenerative diseases, diabetes, arthritis, osteoporosis, etc. The effect of nutraceuticals is similar to pharmaceuticals, even though the term nutraceutical has no regulatory definition. The usage of nutraceuticals, to prevent and treat the aforementioned diseases, is limited by several features such as poor water solubility, low bioavailability, low stability, low permeability, low efficacy, etc. These downsides can be overcome by the application of the field of nanotechnology manipulating the properties and structures of materials at the nanometer scale. In this review, the linear and cyclic dextrin, formed during the enzymatic degradation of starch, are highlighted as highly promising nanomaterials- based drug delivery systems. The modified cyclic dextrin, cyclodextrin (CD)-based nanosponges (NSs), are well-known delivery systems of several nutraceuticals such as quercetin, curcumin, resveratrol, thyme essential oil, melatonin, and appear as a more advanced drug delivery system than modified linear dextrin. CD-based NSs prolong and control the nutraceuticals release, and display higher biocompatibility, stability, and solubility of poorly water-soluble nutraceuticals than the CD-inclusion complexes, or uncomplexed nutraceuticals. In addition, the well-explored CD-based NSs pathways, as drug delivery systems, are described. Although important progress is made in drug delivery, all the findings will serve as a source for the use of CD-based nanosystems for nutraceutical delivery. To sum up, our review introduces the extensive literature about the nutraceutical concepts, synthesis, characterization, and applications of the CD-based nano delivery systems that will further contribute to the nutraceutical delivery with more potent nanosystems based on linear dextrins.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Francesco Trotta
- Department of Chemistry, University of Torino, Via P. Giuria 7, 10125 Torino, Italy; (G.H.); (A.M.); (A.R.P.); (C.C.); (S.L.A.); (Y.K.M.); (F.C.)
| |
Collapse
|
46
|
Circulating tumour cells in the -omics era: how far are we from achieving the 'singularity'? Br J Cancer 2022; 127:173-184. [PMID: 35273384 PMCID: PMC9296521 DOI: 10.1038/s41416-022-01768-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 01/27/2022] [Accepted: 02/17/2022] [Indexed: 12/22/2022] Open
Abstract
Over the past decade, cancer diagnosis has expanded to include liquid biopsies in addition to tissue biopsies. Liquid biopsies can result in earlier and more accurate diagnosis and more effective monitoring of disease progression than tissue biopsies as samples can be collected frequently. Because of these advantages, liquid biopsies are now used extensively in clinical care. Liquid biopsy samples are analysed for circulating tumour cells (CTCs), cell-free DNA, RNA, proteins and exosomes. CTCs originate from the tumour, play crucial roles in metastasis and carry information on tumour heterogeneity. Multiple single-cell omics approaches allow the characterisation of the molecular makeup of CTCs. It has become evident that CTCs are robust biomarkers for predicting therapy response, clinical development of metastasis and disease progression. This review describes CTC biology, molecular heterogeneity within CTCs and the involvement of EMT in CTC dynamics. In addition, we describe the single-cell multi-omics technologies that have provided insights into the molecular features within therapy-resistant and metastasis-prone CTC populations. Functional studies coupled with integrated multi-omics analyses have the potential to identify therapies that can intervene the functions of CTCs.
Collapse
|
47
|
Shi R, Zhang Z, Zhu A, Xiong X, Zhang J, Xu J, Sy MS, Li C. Targeting Type I Collagen for Cancer Treatment. Int J Cancer 2022; 151:665-683. [PMID: 35225360 DOI: 10.1002/ijc.33985] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 02/11/2022] [Accepted: 02/14/2022] [Indexed: 11/07/2022]
Abstract
Collagen is the most abundant protein in animals. Interactions between tumor cells and collagen influence every step of tumor development. Type I collagen is the main fibrillar collagen in the extracellular matrix and is frequently up-regulated during tumorigenesis. The binding of type I collagen to its receptors on tumor cells promotes tumor cell proliferation, epithelial-mesenchymal transition, and metastasis. Type I collagen also regulates the efficacy of tumor therapies, such as chemotherapy, radiotherapy, and immunotherapy. Furthermore, type I collagen fragments are diagnostic markers of metastatic tumors and have prognostic value. Inhibition of type I collagen synthesis has been reported to have anti-tumor effects in animal models. However, collagen has also been shown to possess anti-tumor activity. Therefore, the roles that type I collagen plays in tumor biology are complex and tumor type-dependent. In this review, we discuss the expression and regulation of synthesis of type I collagen, as well as the role up-regulated type I collagen plays in various stages of cancer progression. We also discuss the role of collagen in tumor therapy. Finally, we highlight several recent approaches targeting type I collagen for cancer treatment. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Run Shi
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, Key Laboratory for Cell Homeostasis and Cancer Research of Guangdong High Education Institute, Guangzhou, China
| | - Zhe Zhang
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, Key Laboratory for Cell Homeostasis and Cancer Research of Guangdong High Education Institute, Guangzhou, China
| | - Ankai Zhu
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, Key Laboratory for Cell Homeostasis and Cancer Research of Guangdong High Education Institute, Guangzhou, China
| | - Xingxing Xiong
- Department of Operating Room, Jiangxi Cancer Hospital of Nanchang University, Nanchang, China
| | - Jie Zhang
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, Key Laboratory for Cell Homeostasis and Cancer Research of Guangdong High Education Institute, Guangzhou, China
| | - Jiang Xu
- Department of Stomatology, First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, China
| | - Man-Sun Sy
- Department of Pathology, School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| | - Chaoyang Li
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, Key Laboratory for Cell Homeostasis and Cancer Research of Guangdong High Education Institute, Guangzhou, China
| |
Collapse
|
48
|
Jin Y, Kang Y, Wang M, Wu B, Su B, Yin H, Tang Y, Li Q, Wei W, Mei Q, Hu G, Lukacs-Kornek V, Li J, Wu K, Yuan X, Wang W. Targeting polarized phenotype of microglia via IL6/JAK2/STAT3 signaling to reduce NSCLC brain metastasis. Signal Transduct Target Ther 2022; 7:52. [PMID: 35194016 PMCID: PMC8864012 DOI: 10.1038/s41392-022-00872-9] [Citation(s) in RCA: 66] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 12/29/2021] [Accepted: 12/31/2021] [Indexed: 12/27/2022] Open
Abstract
Tumor-associated macrophages have emerged as crucial factors for metastases. Microglia are indispensable components of the brain microenvironment and play vital roles in brain metastasis (BM). However, the underlying mechanism of how activated microglia promote brain metastasis of non-small cell lung cancer (NSCLC) remains elusive. Here, we purified cell lines with brain-metastatic tropism and employed a co-culture system to reveal their communication with microglia. By single-cell RNA-sequencing and transcriptome difference analysis, we identified IL6 as the key regulator in brain-metastatic cells (A549-F3) to induce anti-inflammatory microglia via JAK2/STAT3 signaling, which in turn promoted the colonization process in metastatic A549-F3 cells. In our clinical samples, patients with higher levels of IL6 in serum showed higher propensity for brain metastasis. Additionally, the TCGA (The Cancer Genome Atlas) data revealed that NSCLC patients with a lower level of IL6 had a longer overall survival time compared to those with a higher level of IL6. Overall, our data indicate that the targeting of IL6/JAK2/STAT3 signaling in activated microglia may be a promising new approach for inhibiting brain metastasis in NSCLC patients.
Collapse
Affiliation(s)
- Yu Jin
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China
| | - Yalin Kang
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China
| | - Minghuan Wang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China
| | - Bili Wu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China
| | - Beibei Su
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China
| | - Han Yin
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China
| | - Yang Tang
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China
| | - Qianxia Li
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China
| | - Wenjie Wei
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China
| | - Qi Mei
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China
| | - Guangyuan Hu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China
| | - Veronika Lukacs-Kornek
- Institute of Experimental Immunology, University Clinic of Rheinische Friedrich-Wilhelms-University, Bonn, Germany
| | - Jian Li
- Institute of Experimental Immunology, University Clinic of Rheinische Friedrich-Wilhelms-University, Bonn, Germany
| | - Kongming Wu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China
| | - Xianglin Yuan
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China.
| | - Wei Wang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China.
| |
Collapse
|
49
|
Henriques-Pons A, Beghini DG, Silva VDS, Iwao Horita S, da Silva FAB. Pulmonary Mesenchymal Stem Cells in Mild Cases of COVID-19 Are Dedicated to Proliferation; In Severe Cases, They Control Inflammation, Make Cell Dispersion, and Tissue Regeneration. Front Immunol 2022; 12:780900. [PMID: 35095855 PMCID: PMC8793136 DOI: 10.3389/fimmu.2021.780900] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 12/17/2021] [Indexed: 12/29/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are multipotent adult stem cells present in virtually all tissues; they have potent self-renewal capacity and differentiate into multiple cell types. For many reasons, these cells are a promising therapeutic alternative to treat patients with severe COVID-19 and pulmonary post-COVID sequelae. These cells are not only essential for tissue regeneration; they can also alter the pulmonary environment through the paracrine secretion of several mediators. They can control or promote inflammation, induce other stem cells differentiation, restrain the virus load, and much more. In this work, we performed single-cell RNA-seq data analysis of MSCs in bronchoalveolar lavage samples from control individuals and COVID-19 patients with mild and severe clinical conditions. When we compared samples from mild cases with control individuals, most genes transcriptionally upregulated in COVID-19 were involved in cell proliferation. However, a new set of genes with distinct biological functions was upregulated when we compared severely affected with mild COVID-19 patients. In this analysis, the cells upregulated genes related to cell dispersion/migration and induced the γ-activated sequence (GAS) genes, probably triggered by IFNGR1 and IFNGR2. Then, IRF-1 was upregulated, one of the GAS target genes, leading to the interferon-stimulated response (ISR) and the overexpression of many signature target genes. The MSCs also upregulated genes involved in the mesenchymal-epithelial transition, virus control, cell chemotaxis, and used the cytoplasmic RNA danger sensors RIG-1, MDA5, and PKR. In a non-comparative analysis, we observed that MSCs from severe cases do not express many NF-κB upstream receptors, such as Toll-like (TLRs) TLR-3, -7, and -8; tumor necrosis factor (TNFR1 or TNFR2), RANK, CD40, and IL-1R1. Indeed, many NF-κB inhibitors were upregulated, including PPP2CB, OPTN, NFKBIA, and FHL2, suggesting that MSCs do not play a role in the "cytokine storm" observed. Therefore, lung MSCs in COVID-19 sense immune danger and act protectively in concert with the pulmonary environment, confirming their therapeutic potential in cell-based therapy for COVID-19. The transcription of MSCs senescence markers is discussed.
Collapse
Affiliation(s)
- Andrea Henriques-Pons
- Fundação Oswaldo Cruz, Instituto Oswaldo Cruz, Laboratório de Inovações em Terapias, Ensino e Bioprodutos, Rio de Janeiro, Brazil
| | - Daniela Gois Beghini
- Fundação Oswaldo Cruz, Instituto Oswaldo Cruz, Laboratório de Inovações em Terapias, Ensino e Bioprodutos, Rio de Janeiro, Brazil
| | | | - Samuel Iwao Horita
- Fundação Oswaldo Cruz, Instituto Oswaldo Cruz, Laboratório de Inovações em Terapias, Ensino e Bioprodutos, Rio de Janeiro, Brazil
| | | |
Collapse
|
50
|
Tanimura K, Yamada T, Okada K, Nakai K, Horinaka M, Katayama Y, Morimoto K, Ogura Y, Takeda T, Shiotsu S, Ichikawa K, Watanabe S, Morimoto Y, Iwasaku M, Kaneko Y, Uchino J, Taniguchi H, Yoneda K, Matoba S, Sakai T, Uehara H, Yano S, Kusaba T, Katayama R, Takayama K. HER3 activation contributes toward the emergence of ALK inhibitor-tolerant cells in ALK-rearranged lung cancer with mesenchymal features. NPJ Precis Oncol 2022; 6:5. [PMID: 35042943 PMCID: PMC8766605 DOI: 10.1038/s41698-021-00250-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Accepted: 12/16/2021] [Indexed: 01/22/2023] Open
Abstract
Anaplastic lymphoma kinase-tyrosine kinase inhibitors (ALK-TKIs) have shown dramatic efficacy in patients with ALK-rearranged lung cancer; however, complete response in these patients is rare. Here, we investigated the molecular mechanisms underlying the emergence and maintenance of drug-tolerant cells in ALK-rearranged lung cancer. Cell based-assays demonstrated that HER3 activation and mesenchymal-to-epithelial transition, mediated through ZEB1 proteins, help maintain cell survival and induce the emergence of ALK-TKI-tolerant cells. Compared with ALK-TKIs alone, cotreatment with pan-HER inhibitor afatinib and ALK-TKIs prevented tumor regrowth, leading to the eradication of tumors in ALK-rearranged tumors with mesenchymal features. Moreover, pre-treatment vimentin expression in clinical specimens obtained from patients with ALK-rearranged lung cancer was associated with poor ALK-TKI treatment outcomes. These results demonstrated that HER3 activation plays a pivotal role in the emergence of ALK-TKI-tolerant cells. Furthermore, the inhibition of HER3 signals combined with ALK-TKIs dramatically improves treatment outcomes for ALK-rearranged lung cancer with mesenchymal features.
Collapse
Affiliation(s)
- Keiko Tanimura
- Department of Pulmonary Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajii-Cho Kawaramachi-Hirokoji, Kamigyo-Ku, Kyoto, 602-8566, Japan
| | - Tadaaki Yamada
- Department of Pulmonary Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajii-Cho Kawaramachi-Hirokoji, Kamigyo-Ku, Kyoto, 602-8566, Japan.
| | - Koutaroh Okada
- Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, 3-8-31, Ariake, Koto, Tokyo, 135-8550, Japan
| | - Kunihiro Nakai
- Department of Nephrology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajii-Cho Kawaramachi-Hirokoji, Kamigyo-Ku, Kyoto, 602-8566, Japan
| | - Mano Horinaka
- Department of Drug Discovery Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajii-Cho Kawaramachi-Hirokoji, Kamigyo-Ku, Kyoto, 602-8566, Japan
| | - Yuki Katayama
- Department of Pulmonary Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajii-Cho Kawaramachi-Hirokoji, Kamigyo-Ku, Kyoto, 602-8566, Japan
| | - Kenji Morimoto
- Department of Pulmonary Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajii-Cho Kawaramachi-Hirokoji, Kamigyo-Ku, Kyoto, 602-8566, Japan
| | - Yuri Ogura
- Department of Respiratory Medicine, Japanese Red Cross Kyoto Daini Hospital, 355-5 Haruobi-Cho, Kamigyo-Ku, Kyoto, 602-8026, Japan
| | - Takayuki Takeda
- Department of Respiratory Medicine, Japanese Red Cross Kyoto Daini Hospital, 355-5 Haruobi-Cho, Kamigyo-Ku, Kyoto, 602-8026, Japan
| | - Shinsuke Shiotsu
- Department of Respiratory Medicine, Japanese Red Cross Kyoto Daiichi Hospital, 15-749, Honmachi, Higashiyama-Ku, Kyoto, 605-0981, Japan
| | - Kosuke Ichikawa
- Department of Respiratory Medicine and Infectious Diseases, Niigata University Graduate School of Medical and Dental Sciences, 2-5274, Gakkocho-Dori, Niigata, 951-8514, Japan
| | - Satoshi Watanabe
- Department of Respiratory Medicine and Infectious Diseases, Niigata University Graduate School of Medical and Dental Sciences, 2-5274, Gakkocho-Dori, Niigata, 951-8514, Japan
| | - Yoshie Morimoto
- Department of Pulmonary Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajii-Cho Kawaramachi-Hirokoji, Kamigyo-Ku, Kyoto, 602-8566, Japan
| | - Masahiro Iwasaku
- Department of Pulmonary Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajii-Cho Kawaramachi-Hirokoji, Kamigyo-Ku, Kyoto, 602-8566, Japan
| | - Yoshiko Kaneko
- Department of Pulmonary Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajii-Cho Kawaramachi-Hirokoji, Kamigyo-Ku, Kyoto, 602-8566, Japan
| | - Junji Uchino
- Department of Pulmonary Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajii-Cho Kawaramachi-Hirokoji, Kamigyo-Ku, Kyoto, 602-8566, Japan
| | - Hirokazu Taniguchi
- Department of Respiratory Medicine, Nagasaki University Graduate School of Biomedical Sciences, 1-12-4, Sakamoto, Nagasaki, 852-8523, Japan
| | - Kazue Yoneda
- University of Occupational and Environmental Health, Second Department of Surgery, 1-1, Iseigaoka, Kitakyushu, Fukuoka, 807-8556, Japan
| | - Satoaki Matoba
- Department of Cardiovascular Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajii-Cho Kawaramachi-Hirokoji, Kamigyo-Ku, Kyoto, 602-8566, Japan
| | - Toshiyuki Sakai
- Department of Drug Discovery Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajii-Cho Kawaramachi-Hirokoji, Kamigyo-Ku, Kyoto, 602-8566, Japan
| | - Hisanori Uehara
- Division of Pathology, Tokushima University Hospital, 2-50-1 Kuramotocho, Tokushima City, Tokushima, 770-8503, Japan
| | - Seiji Yano
- Division of Medical Oncology, Cancer Research Institute, Kanazawa University, Kakuma-Machi, Kanazawa, Ishikaswa, 920-1192, Japan
| | - Tetsuro Kusaba
- Department of Nephrology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajii-Cho Kawaramachi-Hirokoji, Kamigyo-Ku, Kyoto, 602-8566, Japan
| | - Ryohei Katayama
- Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, 3-8-31, Ariake, Koto, Tokyo, 135-8550, Japan
| | - Koichi Takayama
- Department of Pulmonary Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajii-Cho Kawaramachi-Hirokoji, Kamigyo-Ku, Kyoto, 602-8566, Japan
| |
Collapse
|