1
|
Harikumar H, van Royen ME, van Leenders GJLH. 4D pathology: translating dynamic epithelial tubulogenesis to prostate cancer pathology. Histopathology 2025; 86:681-693. [PMID: 39428716 PMCID: PMC11903113 DOI: 10.1111/his.15354] [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] [Indexed: 10/22/2024]
Abstract
The Gleason score is the gold standard for grading of prostate cancer (PCa) and is assessed by assigning specific grades to different microscopical growth patterns. Aside from the Gleason grades, individual growth patterns such as cribriform architecture were recently shown to have independent prognostic value for disease outcome. PCa grading is performed on static tissue samples collected at one point in time, whereas in vivo epithelial tumour structures are dynamically invading, branching and expanding into the surrounding stroma. Due to the lack of models that are able to track human PCa microscopical developments over time, our understanding of underlying tissue dynamics is sparse. We postulate that human PCa expansion utilizes embryonic and developmental tubulogenetic pathways. The aim of this study is to provide a comprehensive overview of developmental pathways of normal epithelial tubule formation, elongation, and branching, and relate those to the static microscopical PCa growth patterns observed in daily clinical practise. This study could provide a rationale for the discerned pathological interobserver variability and the clinical outcome differences between PCa growth patterns.
Collapse
Affiliation(s)
- Hridya Harikumar
- Department of Pathology, Erasmus MC Cancer InstituteUniversity Medical CentreRotterdamthe Netherlands
| | - Martin E van Royen
- Department of Pathology, Erasmus MC Cancer InstituteUniversity Medical CentreRotterdamthe Netherlands
| | - Geert JLH van Leenders
- Department of Pathology, Erasmus MC Cancer InstituteUniversity Medical CentreRotterdamthe Netherlands
| |
Collapse
|
2
|
Alexandrova A, Kontareva E, Pustovalova M, Leonov S, Merkher Y. Navigating the Collective: Nanoparticle-Assisted Identification of Leader Cancer Cells During Migration. Life (Basel) 2025; 15:127. [PMID: 39860067 PMCID: PMC11766853 DOI: 10.3390/life15010127] [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: 11/27/2024] [Revised: 01/11/2025] [Accepted: 01/15/2025] [Indexed: 01/27/2025] Open
Abstract
Cancer-related deaths primarily occur due to metastasis, a process involving the migration and invasion of cancer cells. In most solid tumors, metastasis occurs through collective cell migration (CCM), guided by "cellular leaders". These leader cells generate forces through actomyosin-mediated protrusion and contractility. The cytoskeletal mechanisms employed by metastatic cells during the migration process closely resemble the use of the actin cytoskeleton in endocytosis. In our previous work, we revealed that tumor cells exhibiting high metastatic potential (MP) are more adept at encapsulating 100-200 nm nanoparticles than those with lower MP. The objective of this study was to investigate whether nanoparticle encapsulation could effectively differentiate leader tumor cells during their CCM. To achieve our objectives, we employed a two-dimensional CCM model grounded in the wound-healing ("scratch") assay, utilizing two breast cancer cell lines, MCF7 and MDA-MB-231, which display low and high migratory potential, respectively. We conducted calibration experiments to identify the "optimal time" at which cells exhibit peak speed during wound closure. Furthermore, we carried out experiments to assess nanoparticle uptake, calculating the colocalization coefficient, and employed phalloidin staining to analyze the anisotropy and orientation of actin filaments. The highest activity for low-MP cells was achieved at 2.6 h during the calibration experiments, whereas high-MP cells were maximally active at 3.9 h, resulting in 8% and 11% reductions in wound area, respectively. We observed a significant difference in encapsulation efficiency between leader and peripheral cells for both high-MP (p < 0.013) and low-MP (p < 0.02) cells. Moreover, leader cells demonstrated a considerably higher anisotropy coefficient (p < 0.029), indicating a more organized, directional structure of actin filaments compared to peripheral cells. Thus, nanoparticle encapsulation offers a groundbreaking approach to identifying the most aggressive and invasive leader cells during the CCM process in breast cancer. Detecting these cells is crucial for developing targeted therapies that can effectively curb metastasis and improve patient outcomes.
Collapse
Affiliation(s)
- Anastasia Alexandrova
- The Laboratory of Personalized Chemo-Radiation Therapy, Institute of Future Biophysics, Moscow 141700, Russia; (A.A.); (S.L.)
| | - Elizaveta Kontareva
- The Laboratory of Personalized Chemo-Radiation Therapy, Institute of Future Biophysics, Moscow 141700, Russia; (A.A.); (S.L.)
| | - Margarita Pustovalova
- The Laboratory of Personalized Chemo-Radiation Therapy, Institute of Future Biophysics, Moscow 141700, Russia; (A.A.); (S.L.)
| | - Sergey Leonov
- The Laboratory of Personalized Chemo-Radiation Therapy, Institute of Future Biophysics, Moscow 141700, Russia; (A.A.); (S.L.)
- Institute of Cell Biophysics of Russian Academy of Sciences, Pushchino 142290, Russia
| | - Yulia Merkher
- The Laboratory of Personalized Chemo-Radiation Therapy, Institute of Future Biophysics, Moscow 141700, Russia; (A.A.); (S.L.)
- Faculty of Biomedical Engineering, Technion—Israel Institute of Technology, Haifa 3200003, Israel
| |
Collapse
|
3
|
Bouchareb E, Dallel S, De Haze A, Damon-Soubeyrand C, Renaud Y, Baabdaty E, Vialat M, Fabre J, Pouchin P, De Joussineau C, Degoul F, Sanmukh S, Gendronneau J, Sanchez P, Gonthier-Gueret C, Trousson A, Morel L, Lobaccaro JM, Kocer A, Baron S. Liver X Receptors Enhance Epithelial to Mesenchymal Transition in Metastatic Prostate Cancer Cells. Cancers (Basel) 2024; 16:2776. [PMID: 39199549 PMCID: PMC11353074 DOI: 10.3390/cancers16162776] [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: 06/10/2024] [Revised: 07/12/2024] [Accepted: 07/29/2024] [Indexed: 09/01/2024] Open
Abstract
Prostate cancer (PCa) is one of the most common cancers in men. Metastasis is the leading cause of death in prostate cancer patients. One of the crucial processes involved in metastatic spread is the "epithelial-mesenchymal transition" (EMT), which allows cells to acquire the ability to invade distant organs. Liver X Receptors (LXRs) are nuclear receptors that have been demonstrated to regulate EMT in various cancers, including hepatic cancer. Our study reveals that the LXR pathway can control pro-invasive cell capacities through EMT in prostate cancer, employing ex vivo and in vivo approaches. We characterized the EMT status of the commonly used LNCaP, DU145, and PC3 prostate cancer cell lines through molecular and immunohistochemistry experiments. The impact of LXR activation on EMT function was also assessed by analyzing the migration and invasion of these cell lines in the absence or presence of an LXR agonist. Using in vivo experiments involving NSG-immunodeficient mice xenografted with PC3-GFP cells, we were able to study metastatic spread and the effect of LXRs on this process. LXR activation led to an increase in the accumulation of Vimentin and Amphiregulin in PC3. Furthermore, the migration of PC3 cells significantly increased in the presence of the LXR agonist, correlating with an upregulation of EMT. Interestingly, LXR activation significantly increased metastatic spread in an NSG mouse model. Overall, this work identifies a promoting effect of LXRs on EMT in the PC3 model of advanced prostate cancer.
Collapse
Affiliation(s)
- Erwan Bouchareb
- iGReD, CNRS UMR 6293, INSERM U1103, Université Clermont Auvergne, 28, Place Henri Dunant, BP38, 63001 Clermont-Ferrand, France; (E.B.); (S.D.); (C.D.-S.); (Y.R.); (E.B.); (M.V.); (J.F.); (P.P.); (C.D.J.); (F.D.); (S.S.); (J.G.); (P.S.); (C.G.-G.); (A.T.); (L.M.); (J.M.L.)
- Groupe Cancer Clermont Auvergne, 28, Place Henri Dunant, BP38, 63001 Clermont-Ferrand, France
| | - Sarah Dallel
- iGReD, CNRS UMR 6293, INSERM U1103, Université Clermont Auvergne, 28, Place Henri Dunant, BP38, 63001 Clermont-Ferrand, France; (E.B.); (S.D.); (C.D.-S.); (Y.R.); (E.B.); (M.V.); (J.F.); (P.P.); (C.D.J.); (F.D.); (S.S.); (J.G.); (P.S.); (C.G.-G.); (A.T.); (L.M.); (J.M.L.)
- Groupe Cancer Clermont Auvergne, 28, Place Henri Dunant, BP38, 63001 Clermont-Ferrand, France
- Service d’Endocrinologie, Diabétologie et Maladies Métaboliques, CHU Clermont Ferrand, Hôpital Gabriel Montpied, 63003 Clermont-Ferrand, France
| | - Angélique De Haze
- iGReD, CNRS UMR 6293, INSERM U1103, Université Clermont Auvergne, 28, Place Henri Dunant, BP38, 63001 Clermont-Ferrand, France; (E.B.); (S.D.); (C.D.-S.); (Y.R.); (E.B.); (M.V.); (J.F.); (P.P.); (C.D.J.); (F.D.); (S.S.); (J.G.); (P.S.); (C.G.-G.); (A.T.); (L.M.); (J.M.L.)
- Groupe Cancer Clermont Auvergne, 28, Place Henri Dunant, BP38, 63001 Clermont-Ferrand, France
| | - Christelle Damon-Soubeyrand
- iGReD, CNRS UMR 6293, INSERM U1103, Université Clermont Auvergne, 28, Place Henri Dunant, BP38, 63001 Clermont-Ferrand, France; (E.B.); (S.D.); (C.D.-S.); (Y.R.); (E.B.); (M.V.); (J.F.); (P.P.); (C.D.J.); (F.D.); (S.S.); (J.G.); (P.S.); (C.G.-G.); (A.T.); (L.M.); (J.M.L.)
- Groupe Cancer Clermont Auvergne, 28, Place Henri Dunant, BP38, 63001 Clermont-Ferrand, France
| | - Yoan Renaud
- iGReD, CNRS UMR 6293, INSERM U1103, Université Clermont Auvergne, 28, Place Henri Dunant, BP38, 63001 Clermont-Ferrand, France; (E.B.); (S.D.); (C.D.-S.); (Y.R.); (E.B.); (M.V.); (J.F.); (P.P.); (C.D.J.); (F.D.); (S.S.); (J.G.); (P.S.); (C.G.-G.); (A.T.); (L.M.); (J.M.L.)
- Groupe Cancer Clermont Auvergne, 28, Place Henri Dunant, BP38, 63001 Clermont-Ferrand, France
| | - Elissa Baabdaty
- iGReD, CNRS UMR 6293, INSERM U1103, Université Clermont Auvergne, 28, Place Henri Dunant, BP38, 63001 Clermont-Ferrand, France; (E.B.); (S.D.); (C.D.-S.); (Y.R.); (E.B.); (M.V.); (J.F.); (P.P.); (C.D.J.); (F.D.); (S.S.); (J.G.); (P.S.); (C.G.-G.); (A.T.); (L.M.); (J.M.L.)
- Groupe Cancer Clermont Auvergne, 28, Place Henri Dunant, BP38, 63001 Clermont-Ferrand, France
| | - Marine Vialat
- iGReD, CNRS UMR 6293, INSERM U1103, Université Clermont Auvergne, 28, Place Henri Dunant, BP38, 63001 Clermont-Ferrand, France; (E.B.); (S.D.); (C.D.-S.); (Y.R.); (E.B.); (M.V.); (J.F.); (P.P.); (C.D.J.); (F.D.); (S.S.); (J.G.); (P.S.); (C.G.-G.); (A.T.); (L.M.); (J.M.L.)
- Groupe Cancer Clermont Auvergne, 28, Place Henri Dunant, BP38, 63001 Clermont-Ferrand, France
| | - Julien Fabre
- iGReD, CNRS UMR 6293, INSERM U1103, Université Clermont Auvergne, 28, Place Henri Dunant, BP38, 63001 Clermont-Ferrand, France; (E.B.); (S.D.); (C.D.-S.); (Y.R.); (E.B.); (M.V.); (J.F.); (P.P.); (C.D.J.); (F.D.); (S.S.); (J.G.); (P.S.); (C.G.-G.); (A.T.); (L.M.); (J.M.L.)
- Groupe Cancer Clermont Auvergne, 28, Place Henri Dunant, BP38, 63001 Clermont-Ferrand, France
| | - Pierre Pouchin
- iGReD, CNRS UMR 6293, INSERM U1103, Université Clermont Auvergne, 28, Place Henri Dunant, BP38, 63001 Clermont-Ferrand, France; (E.B.); (S.D.); (C.D.-S.); (Y.R.); (E.B.); (M.V.); (J.F.); (P.P.); (C.D.J.); (F.D.); (S.S.); (J.G.); (P.S.); (C.G.-G.); (A.T.); (L.M.); (J.M.L.)
- Groupe Cancer Clermont Auvergne, 28, Place Henri Dunant, BP38, 63001 Clermont-Ferrand, France
| | - Cyrille De Joussineau
- iGReD, CNRS UMR 6293, INSERM U1103, Université Clermont Auvergne, 28, Place Henri Dunant, BP38, 63001 Clermont-Ferrand, France; (E.B.); (S.D.); (C.D.-S.); (Y.R.); (E.B.); (M.V.); (J.F.); (P.P.); (C.D.J.); (F.D.); (S.S.); (J.G.); (P.S.); (C.G.-G.); (A.T.); (L.M.); (J.M.L.)
- Groupe Cancer Clermont Auvergne, 28, Place Henri Dunant, BP38, 63001 Clermont-Ferrand, France
| | - Françoise Degoul
- iGReD, CNRS UMR 6293, INSERM U1103, Université Clermont Auvergne, 28, Place Henri Dunant, BP38, 63001 Clermont-Ferrand, France; (E.B.); (S.D.); (C.D.-S.); (Y.R.); (E.B.); (M.V.); (J.F.); (P.P.); (C.D.J.); (F.D.); (S.S.); (J.G.); (P.S.); (C.G.-G.); (A.T.); (L.M.); (J.M.L.)
- Groupe Cancer Clermont Auvergne, 28, Place Henri Dunant, BP38, 63001 Clermont-Ferrand, France
| | - Swapnil Sanmukh
- iGReD, CNRS UMR 6293, INSERM U1103, Université Clermont Auvergne, 28, Place Henri Dunant, BP38, 63001 Clermont-Ferrand, France; (E.B.); (S.D.); (C.D.-S.); (Y.R.); (E.B.); (M.V.); (J.F.); (P.P.); (C.D.J.); (F.D.); (S.S.); (J.G.); (P.S.); (C.G.-G.); (A.T.); (L.M.); (J.M.L.)
- Groupe Cancer Clermont Auvergne, 28, Place Henri Dunant, BP38, 63001 Clermont-Ferrand, France
| | - Juliette Gendronneau
- iGReD, CNRS UMR 6293, INSERM U1103, Université Clermont Auvergne, 28, Place Henri Dunant, BP38, 63001 Clermont-Ferrand, France; (E.B.); (S.D.); (C.D.-S.); (Y.R.); (E.B.); (M.V.); (J.F.); (P.P.); (C.D.J.); (F.D.); (S.S.); (J.G.); (P.S.); (C.G.-G.); (A.T.); (L.M.); (J.M.L.)
- Groupe Cancer Clermont Auvergne, 28, Place Henri Dunant, BP38, 63001 Clermont-Ferrand, France
| | - Phelipe Sanchez
- iGReD, CNRS UMR 6293, INSERM U1103, Université Clermont Auvergne, 28, Place Henri Dunant, BP38, 63001 Clermont-Ferrand, France; (E.B.); (S.D.); (C.D.-S.); (Y.R.); (E.B.); (M.V.); (J.F.); (P.P.); (C.D.J.); (F.D.); (S.S.); (J.G.); (P.S.); (C.G.-G.); (A.T.); (L.M.); (J.M.L.)
- Groupe Cancer Clermont Auvergne, 28, Place Henri Dunant, BP38, 63001 Clermont-Ferrand, France
| | - Céline Gonthier-Gueret
- iGReD, CNRS UMR 6293, INSERM U1103, Université Clermont Auvergne, 28, Place Henri Dunant, BP38, 63001 Clermont-Ferrand, France; (E.B.); (S.D.); (C.D.-S.); (Y.R.); (E.B.); (M.V.); (J.F.); (P.P.); (C.D.J.); (F.D.); (S.S.); (J.G.); (P.S.); (C.G.-G.); (A.T.); (L.M.); (J.M.L.)
- Groupe Cancer Clermont Auvergne, 28, Place Henri Dunant, BP38, 63001 Clermont-Ferrand, France
| | - Amalia Trousson
- iGReD, CNRS UMR 6293, INSERM U1103, Université Clermont Auvergne, 28, Place Henri Dunant, BP38, 63001 Clermont-Ferrand, France; (E.B.); (S.D.); (C.D.-S.); (Y.R.); (E.B.); (M.V.); (J.F.); (P.P.); (C.D.J.); (F.D.); (S.S.); (J.G.); (P.S.); (C.G.-G.); (A.T.); (L.M.); (J.M.L.)
- Groupe Cancer Clermont Auvergne, 28, Place Henri Dunant, BP38, 63001 Clermont-Ferrand, France
| | - Laurent Morel
- iGReD, CNRS UMR 6293, INSERM U1103, Université Clermont Auvergne, 28, Place Henri Dunant, BP38, 63001 Clermont-Ferrand, France; (E.B.); (S.D.); (C.D.-S.); (Y.R.); (E.B.); (M.V.); (J.F.); (P.P.); (C.D.J.); (F.D.); (S.S.); (J.G.); (P.S.); (C.G.-G.); (A.T.); (L.M.); (J.M.L.)
- Groupe Cancer Clermont Auvergne, 28, Place Henri Dunant, BP38, 63001 Clermont-Ferrand, France
| | - Jean Marc Lobaccaro
- iGReD, CNRS UMR 6293, INSERM U1103, Université Clermont Auvergne, 28, Place Henri Dunant, BP38, 63001 Clermont-Ferrand, France; (E.B.); (S.D.); (C.D.-S.); (Y.R.); (E.B.); (M.V.); (J.F.); (P.P.); (C.D.J.); (F.D.); (S.S.); (J.G.); (P.S.); (C.G.-G.); (A.T.); (L.M.); (J.M.L.)
- Groupe Cancer Clermont Auvergne, 28, Place Henri Dunant, BP38, 63001 Clermont-Ferrand, France
| | - Ayhan Kocer
- iGReD, CNRS UMR 6293, INSERM U1103, Université Clermont Auvergne, 28, Place Henri Dunant, BP38, 63001 Clermont-Ferrand, France; (E.B.); (S.D.); (C.D.-S.); (Y.R.); (E.B.); (M.V.); (J.F.); (P.P.); (C.D.J.); (F.D.); (S.S.); (J.G.); (P.S.); (C.G.-G.); (A.T.); (L.M.); (J.M.L.)
- Groupe Cancer Clermont Auvergne, 28, Place Henri Dunant, BP38, 63001 Clermont-Ferrand, France
| | - Silvère Baron
- iGReD, CNRS UMR 6293, INSERM U1103, Université Clermont Auvergne, 28, Place Henri Dunant, BP38, 63001 Clermont-Ferrand, France; (E.B.); (S.D.); (C.D.-S.); (Y.R.); (E.B.); (M.V.); (J.F.); (P.P.); (C.D.J.); (F.D.); (S.S.); (J.G.); (P.S.); (C.G.-G.); (A.T.); (L.M.); (J.M.L.)
- Groupe Cancer Clermont Auvergne, 28, Place Henri Dunant, BP38, 63001 Clermont-Ferrand, France
| |
Collapse
|
4
|
Morales-Camilo N, Liu J, Ramírez MJ, Canales-Salgado P, Alegría JJ, Liu X, Ong HT, Barrera NP, Fierro A, Toyama Y, Goult BT, Wang Y, Meng Y, Nishimura R, Fong-Ngern K, Low CSL, Kanchanawong P, Yan J, Ravasio A, Bertocchi C. Alternative molecular mechanisms for force transmission at adherens junctions via β-catenin-vinculin interaction. Nat Commun 2024; 15:5608. [PMID: 38969637 PMCID: PMC11226457 DOI: 10.1038/s41467-024-49850-5] [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: 09/19/2023] [Accepted: 06/21/2024] [Indexed: 07/07/2024] Open
Abstract
Force transmission through adherens junctions (AJs) is crucial for multicellular organization, wound healing and tissue regeneration. Recent studies shed light on the molecular mechanisms of mechanotransduction at the AJs. However, the canonical model fails to explain force transmission when essential proteins of the mechanotransduction module are mutated or missing. Here, we demonstrate that, in absence of α-catenin, β-catenin can directly and functionally interact with vinculin in its open conformation, bearing physiological forces. Furthermore, we found that β-catenin can prevent vinculin autoinhibition in the presence of α-catenin by occupying vinculin´s head-tail interaction site, thus preserving force transmission capability. Taken together, our findings suggest a multi-step force transmission process at AJs, where α-catenin and β-catenin can alternatively and cooperatively interact with vinculin. This can explain the graded responses needed to maintain tissue mechanical homeostasis and, importantly, unveils a force-bearing mechanism involving β-catenin and extended vinculin that can potentially explain the underlying process enabling collective invasion of metastatic cells lacking α-catenin.
Collapse
Affiliation(s)
- Nicole Morales-Camilo
- Laboratory for Molecular Mechanics of Cell Adhesion, Faculty of Biological Sciences, Pontificia Universidad Católica De Chile, Santiago, Chile
| | - Jingzhun Liu
- Department of Physics, National University of Singapore, 117542, Singapore, Singapore
| | - Manuel J Ramírez
- Laboratory for Molecular Mechanics of Cell Adhesion, Faculty of Biological Sciences, Pontificia Universidad Católica De Chile, Santiago, Chile
- Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Patricio Canales-Salgado
- Institute for Biological and Medical Engineering, Schools of Engineering, Medicine and Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
- Faculty of Medical Sciences, Universidad de Santiago de Chile, Santiago, Chile
| | - Juan José Alegría
- Institute for Biological and Medical Engineering, Schools of Engineering, Medicine and Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
- Millennium Institute for Foundational Research on Data (IMFD), Santiago, Chile
| | - Xuyao Liu
- Department of Physics, National University of Singapore, 117542, Singapore, Singapore
- Mechanobiology Institute, Singapore, National University of Singapore, 117411, Singapore, Singapore
| | - Hui Ting Ong
- Mechanobiology Institute, Singapore, National University of Singapore, 117411, Singapore, Singapore
| | - Nelson P Barrera
- Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Angélica Fierro
- Department of Organic Chemistry, School of Chemistry, Faculty of Chemistry and Pharmacy, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Yusuke Toyama
- Mechanobiology Institute, Singapore, National University of Singapore, 117411, Singapore, Singapore
- Department of Biological Sciences, National University of Singapore, Singapore, 117543, Singapore
| | - Benjamin T Goult
- School of Biosciences, University of Kent, Kent, Canterbury, CT2 7NJ, UK
- Department of Biochemistry, Cell & Systems Biology, Institute of Systems, Molecular & Integrative Biology, University of Liverpool, Crown Street, Liverpool, L69 7ZB, UK
| | - Yilin Wang
- Mechanobiology Institute, Singapore, National University of Singapore, 117411, Singapore, Singapore
| | - Yue Meng
- Mechanobiology Institute, Singapore, National University of Singapore, 117411, Singapore, Singapore
| | - Ryosuke Nishimura
- Mechanobiology Institute, Singapore, National University of Singapore, 117411, Singapore, Singapore
| | - Kedsarin Fong-Ngern
- Mechanobiology Institute, Singapore, National University of Singapore, 117411, Singapore, Singapore
| | - Christine Siok Lan Low
- Mechanobiology Institute, Singapore, National University of Singapore, 117411, Singapore, Singapore
| | - Pakorn Kanchanawong
- Mechanobiology Institute, Singapore, National University of Singapore, 117411, Singapore, Singapore
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, 117543, Singapore, Singapore
| | - Jie Yan
- Department of Physics, National University of Singapore, 117542, Singapore, Singapore
- Mechanobiology Institute, Singapore, National University of Singapore, 117411, Singapore, Singapore
| | - Andrea Ravasio
- Institute for Biological and Medical Engineering, Schools of Engineering, Medicine and Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile.
| | - Cristina Bertocchi
- Laboratory for Molecular Mechanics of Cell Adhesion, Faculty of Biological Sciences, Pontificia Universidad Católica De Chile, Santiago, Chile.
- Graduate School of Engineering Science, Osaka University, Osaka, Japan.
| |
Collapse
|
5
|
Ditto M, Jacho D, Eisenmann KM, Yildirim-Ayan E. Extracellular Mechanical Stimuli Alters the Metastatic Progression of Prostate Cancer Cells within 3D Tissue Matrix. Bioengineering (Basel) 2023; 10:1271. [PMID: 38002395 PMCID: PMC10669840 DOI: 10.3390/bioengineering10111271] [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: 09/28/2023] [Revised: 10/19/2023] [Accepted: 10/24/2023] [Indexed: 11/26/2023] Open
Abstract
This study aimed to understand extracellular mechanical stimuli's effect on prostate cancer cells' metastatic progression within a three-dimensional (3D) bone-like microenvironment. In this study, a mechanical loading platform, EQUicycler, has been employed to create physiologically relevant static and cyclic mechanical stimuli to a prostate cancer cell (PC-3)-embedded 3D tissue matrix. Three mechanical stimuli conditions were applied: control (no loading), cyclic (1% strain at 1 Hz), and static mechanical stimuli (1% strain). The changes in prostate cancer cells' cytoskeletal reorganization, polarity (elongation index), proliferation, expression level of N-Cadherin (metastasis-associated gene), and migratory potential within the 3D collagen structures were assessed upon mechanical stimuli. The results have shown that static mechanical stimuli increased the metastasis progression factors, including cell elongation (p < 0.001), cellular F-actin accumulation (p < 0.001), actin polymerization (p < 0.001), N-Cadherin gene expression, and invasion capacity of PC-3 cells within a bone-like microenvironment compared to its cyclic and control loading counterparts. This study established a novel system for studying metastatic cancer cells within bone and enables the creation of biomimetic in vitro models for cancer research and mechanobiology.
Collapse
Affiliation(s)
- Maggie Ditto
- Department of Bioengineering, College of Engineering, University of Toledo, Toledo, OH 43606, USA
| | - Diego Jacho
- Department of Bioengineering, College of Engineering, University of Toledo, Toledo, OH 43606, USA
| | - Kathryn M. Eisenmann
- Department of Cell and Cancer Biology, College of Medicine and Life Sciences, University of Toledo Health Science Campus, Toledo, OH 43614, USA
| | - Eda Yildirim-Ayan
- Department of Bioengineering, College of Engineering, University of Toledo, Toledo, OH 43606, USA
| |
Collapse
|
6
|
Quan Y, Ping H, Wang M, Zhang X. RNA-Sequencing Analysis Indicates That N-Cadherin Promotes Prostate Cancer Progression by the Epigenetic Modification of Key Genes. DNA Cell Biol 2023; 42:563-577. [PMID: 37540080 DOI: 10.1089/dna.2023.0089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/05/2023] Open
Abstract
N-cadherin (cadherin-2 [CDH2]) is widely known as the promoter of prostate cancer (PCa) invasion and castration resistance. However, the biological mechanism of N-cadherin in PCa progression is unclear. In this study, we overexpressed N-cadherin in LNCaP cells and downregulated N-cadherin in PC3 cells by lentiviral transduction. Then, differentially expressed genes (DEGs) and dysregulated biological functions were investigated through RNA sequencing (RNA-seq) analyses. We found 13 long noncoding RNA (lncRNA) transcripts, 72 messenger RNA (mRNA) transcripts, and 3 integrated genes were dysregulated by N-cadherin. In the disease enrichment, bone cancer, and neurodegenerative and nervous system diseases were associated with N-cadherin in the circular RNA (circRNA; PC3 versus [vs.,/] LNCaP [PC3/LNCaP] comparison) and DEG analysis (LNCaP_oe_CDH2 vs. LNCaP_oe_NC [LNCaP_oe_CDH2/NC] comparison). Epigenetic reprogramming, such as nucleic acid binding, and chromatin and histone modifications, was enriched in Gene Ontology (GO) analysis (DEGs in LNCaP_oe_CDH2/NC and PC3_sh_NC/CDH2, and host genes of circRNA in PC3/LNCaP). Transcriptional misregulation in cancer, post-translational protein modification, gene expression, and generic transcription pathways were dysregulated in the pathway enrichment analysis (host genes of circRNA in PC3/LNCaP, and DEGs in LNCaP_oe_CDH2/NC and PC3_sh_NC/CDH2). Verifying DEGs through TCGA-PRAD dataset revealed six oncogenes (ARHGEF1, GRAMD1A, GTF2H4, MAPK8IP3, POLD1, and PTBP1) that were commonly upregulated by N-cadherin and in advanced PCa stages. In summary, we identified several oncogenes and biological functions associated with N-cadherin expression in PCa cells. N-cadherin may trigger epigenetic reprogramming in PCa cells to promote tumor progression.
Collapse
Affiliation(s)
- Yongjun Quan
- Department of Urology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Hao Ping
- Department of Urology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Mingdong Wang
- Department of Urology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Xiaodong Zhang
- Department of Urology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
7
|
Sharma R, Balta S, Raza A, Escalona RM, Kannourakis G, Prithviraj P, Ahmed N. In Vitro and In Silico Analysis of Epithelial-Mesenchymal Transition and Cancer Stemness as Prognostic Markers of Clear Cell Renal Cell Carcinoma. Cancers (Basel) 2023; 15:cancers15092586. [PMID: 37174052 PMCID: PMC10177434 DOI: 10.3390/cancers15092586] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 04/27/2023] [Accepted: 04/28/2023] [Indexed: 05/15/2023] Open
Abstract
The process of epithelial-mesenchymal transition (EMT) involves the phenotypic transformation of cells from epithelial to mesenchymal status. The cells exhibiting EMT contain features of cancer stem cells (CSC), and the dual processes are responsible for progressive cancers. Activation of hypoxia-inducible factors (HIF) is fundamental to the pathogenesis of clear cell renal cell carcinoma (ccRCC), and their role in promoting EMT and CSCs is crucial for ccRCC tumour cell survival, disease progression, and metastatic spread. In this study, we explored the status of HIF genes and their downstream targets, EMT and CSC markers, by immunohistochemistry on in-house accrued ccRCC biopsies and adjacent non-tumorous tissues from patients undergoing partial or radical nephrectomy. In combination, we comprehensively analysed the expression of HIF genes and its downstream EMT and CSC-associated targets relevant to ccRCC by using publicly available datasets, the cancer genome atlas (TCGA) and the clinical proteome tumour analysis consortium (CPTAC). The aim was to search for novel biological prognostic markers that can stratify high-risk patients likely to experience metastatic disease. Using the above two approaches, we report the development of novel gene signatures that may help to identify patients at a high risk of developing metastatic and progressive disease.
Collapse
Affiliation(s)
- Revati Sharma
- Fiona Elsey Cancer Research Institute, Ballarat, VIC 3353, Australia
- Health Innovation and Transformation Centre, Mt Helen Campus, Federation University Australia, Ballarat, VIC 3350, Australia
| | - Showan Balta
- Dorevitch Pathology, Ballarat Base Hospital, Drummond Street, Ballarat, VIC 3350, Australia
| | - Ali Raza
- Fiona Elsey Cancer Research Institute, Ballarat, VIC 3353, Australia
- Health Innovation and Transformation Centre, Mt Helen Campus, Federation University Australia, Ballarat, VIC 3350, Australia
| | - Ruth M Escalona
- Fiona Elsey Cancer Research Institute, Ballarat, VIC 3353, Australia
- Centre for Reproductive Health, The Hudson Institute of Medical Research, 27-31 Wright Street, Clayton, VIC 3168, Australia
- Department of Molecular and Translational Science, Monash University, Clayton, VIC 3168, Australia
| | - George Kannourakis
- Fiona Elsey Cancer Research Institute, Ballarat, VIC 3353, Australia
- Health Innovation and Transformation Centre, Mt Helen Campus, Federation University Australia, Ballarat, VIC 3350, Australia
| | - Prashanth Prithviraj
- Fiona Elsey Cancer Research Institute, Ballarat, VIC 3353, Australia
- Health Innovation and Transformation Centre, Mt Helen Campus, Federation University Australia, Ballarat, VIC 3350, Australia
| | - Nuzhat Ahmed
- Fiona Elsey Cancer Research Institute, Ballarat, VIC 3353, Australia
- Health Innovation and Transformation Centre, Mt Helen Campus, Federation University Australia, Ballarat, VIC 3350, Australia
- Centre for Reproductive Health, The Hudson Institute of Medical Research, 27-31 Wright Street, Clayton, VIC 3168, Australia
- Department of Molecular and Translational Science, Monash University, Clayton, VIC 3168, Australia
- Department of Obstetrics and Gynaecology, University of Melbourne, Parkville, VIC 3010, Australia
| |
Collapse
|
8
|
Archer Goode E, Wang N, Munkley J. Prostate cancer bone metastases biology and clinical management (Review). Oncol Lett 2023; 25:163. [PMID: 36960185 PMCID: PMC10028493 DOI: 10.3892/ol.2023.13749] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 02/09/2023] [Indexed: 03/25/2023] Open
Abstract
Prostate cancer (PCa) is one of the most prominent causes of cancer-related mortality in the male population. A highly impactful prognostic factor for patients diagnosed with PCa is the presence or absence of bone metastases. The formation of secondary tumours at the bone is the most commonly observed site for the establishment of PCa metastases and is associated with reduced survival of patients in addition to a cohort of life-debilitating symptoms, including mobility issues and chronic pain. Despite the prevalence of this disease presentation and the high medical relevance of bone metastases, the mechanisms underlying the formation of metastases to the bone and the understanding of what drives the osteotropism exhibited by prostate tumours remain to be fully elucidated. This lack of in-depth understanding manifests in limited effective treatment options for patients with advanced metastatic PCa and culminates in the low rate of survival observed for this sub-set of patients. The present review aims to summarise the most recent promising advances in the understanding of how and why prostate tumours metastasise to the bone, with the ultimate aim of highlighting novel treatment and prognostic targets, which may provide the opportunity to improve the diagnosis and treatment of patients with PCa with bone metastases.
Collapse
Affiliation(s)
- Emily Archer Goode
- Newcastle University Centre for Cancer, Newcastle University Institute of Biosciences, International Centre for Life, Newcastle NE1 3BZ, UK
| | - Ning Wang
- The Mellanby Centre for Musculoskeletal Research, Department of Oncology and Metabolism, The University of Sheffield, Sheffield S10 2RX, UK
| | - Jennifer Munkley
- Newcastle University Centre for Cancer, Newcastle University Institute of Biosciences, International Centre for Life, Newcastle NE1 3BZ, UK
| |
Collapse
|
9
|
Ritch SJ, Noman ASM, Goyeneche AA, Telleria CM. The metastatic capacity of high-grade serous ovarian cancer cells changes along disease progression: inhibition by mifepristone. Cancer Cell Int 2022; 22:397. [PMID: 36494669 PMCID: PMC9733158 DOI: 10.1186/s12935-022-02822-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 12/03/2022] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Simplistic two-dimensional (2D) in vitro assays have long been the standard for studying the metastatic abilities of cancer cells. However, tri-dimensional (3D) organotypic models provide a more complex environment, closer to that seen in patients, and thereby provide a more accurate representation of their true capabilities. Our laboratory has previously shown that the antiprogestin and antiglucocorticoid mifepristone can reduce the growth, adhesion, migration, and invasion of various aggressive cancer cells assessed using 2D assays. In this study, we characterize the metastatic capabilities of high-grade serous ovarian cancer cells generated along disease progression, in both 2D and 3D assays, and the ability of cytostatic doses of mifepristone to inhibit them. METHODS High-grade serous ovarian cancer cells collected from two separate patients at different stages of their disease were used throughout the study. The 2D wound healing and Boyden chamber assays were used to study migration, while a layer of extracellular matrix was added to the Boyden chamber to study invasion. A 3D organotypic model, composed of fibroblasts embedded in collagen I and topped with a monolayer of mesothelial cells was used to further study cancer cell adhesion and mesothelial displacement. All assays were studied in cells, which were originally harvested from two patients at different stages of disease progression, in the absence or presence of cytostatic doses of mifepristone. RESULTS 2D in vitro assays demonstrated that the migration and invasive rates of the cells isolated from both patients decreased along disease progression. Conversely, in both patients, cells representing late-stage disease demonstrated a higher adhesion capacity to the 3D organotypic model than those representing an early-stage disease. This adhesive behavior is associated with the in vivo tumor capacity of the cells. Regardless of these differences in adhesive, migratory, and invasive behavior among the experimental protocols used, cytostatic doses of mifepristone were able to inhibit the adhesion, migration, and invasion rates of all cells studied, regardless of their basal capabilities over simplistic or organotypic metastatic in vitro model systems. Finally, we demonstrate that when cells acquire the capacity to grow spontaneously as spheroids, they do attach to a 3D organotypic model system when pre-incubated with conditioned media. Of relevance, mifepristone was able to cause dissociation of these multicellular structures. CONCLUSION Differences in cellular behaviours were observed between 2 and 3D assays when studying the metastatic capabilities of high-grade serous ovarian cancer cells representing disease progression. Mifepristone inhibited these metastatic capabilities in all assays studied.
Collapse
Affiliation(s)
- Sabrina J. Ritch
- grid.14709.3b0000 0004 1936 8649Experimental Pathology Unit, Department of Pathology, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC Canada
| | - Abu Shadat M. Noman
- grid.413089.70000 0000 9744 3393Department of Biochemistry and Molecular Biology, Chittagong University, Chittagong, Bangladesh
| | - Alicia A. Goyeneche
- grid.14709.3b0000 0004 1936 8649Experimental Pathology Unit, Department of Pathology, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC Canada ,grid.63984.300000 0000 9064 4811Cancer Research Program, Research Institute, McGill University Health Centre, Montreal, QC Canada
| | - Carlos M. Telleria
- grid.14709.3b0000 0004 1936 8649Experimental Pathology Unit, Department of Pathology, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC Canada ,grid.63984.300000 0000 9064 4811Cancer Research Program, Research Institute, McGill University Health Centre, Montreal, QC Canada
| |
Collapse
|
10
|
Molter CW, Muszynski EF, Tao Y, Trivedi T, Clouvel A, Ehrlicher AJ. Prostate cancer cells of increasing metastatic potential exhibit diverse contractile forces, cell stiffness, and motility in a microenvironment stiffness-dependent manner. Front Cell Dev Biol 2022; 10:932510. [PMID: 36200037 PMCID: PMC9527313 DOI: 10.3389/fcell.2022.932510] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 08/23/2022] [Indexed: 11/13/2022] Open
Abstract
During metastasis, all cancer types must migrate through crowded multicellular environments. Simultaneously, cancers appear to change their biophysical properties. Indeed, cell softening and increased contractility are emerging as seemingly ubiquitous biomarkers of metastatic progression which may facilitate metastasis. Cell stiffness and contractility are also influenced by the microenvironment. Stiffer matrices resembling the tumor microenvironment cause metastatic cells to contract more strongly, further promoting contractile tumorigenic phenotypes. Prostate cancer (PCa), however, appears to deviate from these common cancer biophysics trends; aggressive metastatic PCa cells appear stiffer, rather than softer, to their lowly metastatic PCa counterparts. Although metastatic PCa cells have been reported to be more contractile than healthy cells, how cell contractility changes with increasing PCa metastatic potential has remained unknown. Here, we characterize the biophysical changes of PCa cells of various metastatic potential as a function of microenvironment stiffness. Using a panel of progressively increasing metastatic potential cell lines (22RV1, LNCaP, DU145, and PC3), we quantified their contractility using traction force microscopy (TFM), and measured their cortical stiffness using optical magnetic twisting cytometry (OMTC) and their motility using time-lapse microscopy. We found that PCa contractility, cell stiffness, and motility do not universally scale with metastatic potential. Rather, PCa cells of various metastatic efficiencies exhibit unique biophysical responses that are differentially influenced by substrate stiffness. Despite this biophysical diversity, this work concludes that mechanical microenvironment is a key determinant in the biophysical response of PCa with variable metastatic potentials. The mechanics-oriented focus and methodology of the study is unique and complementary to conventional biochemical and genetic strategies typically used to understand this disease, and thus may usher in new perspectives and approaches.
Collapse
Affiliation(s)
- Clayton W. Molter
- Department of Bioengineering, McGill University, Montreal, QC, Canada
| | - Eliana F. Muszynski
- Department of Bioengineering, McGill University, Montreal, QC, Canada
- Department of Neuroscience, McGill University, Montreal, QC, Canada
| | - Yuanyuan Tao
- Department of Bioengineering, McGill University, Montreal, QC, Canada
- Department of Electrical and Computer Engineering, McGill University, Montreal, QC, Canada
| | - Tanisha Trivedi
- Department of Bioengineering, McGill University, Montreal, QC, Canada
- Department of Anatomy and Cell Biology, McGill University, Montreal, QC, Canada
| | - Anna Clouvel
- Department of Bioengineering, McGill University, Montreal, QC, Canada
| | - Allen J. Ehrlicher
- Department of Bioengineering, McGill University, Montreal, QC, Canada
- Department of Anatomy and Cell Biology, McGill University, Montreal, QC, Canada
- Rosalind and Morris Goodman Cancer Research Institute, McGill University, Montreal, QC, Canada
- Department of Biomedical Engineering, McGill University, Montreal, QC, Canada
- Department of Mechanical Engineering, McGill University, Montreal, QC, Canada
| |
Collapse
|
11
|
Karnas K, Strączek T, Kapusta C, Lekka M, Dulińska-Litewka J, Karewicz A. Specific Binding of Novel SPION-Based System Bearing Anti-N-Cadherin Antibodies to Prostate Tumor Cells. Int J Nanomedicine 2021; 16:6537-6552. [PMID: 34602817 PMCID: PMC8478793 DOI: 10.2147/ijn.s324354] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 08/30/2021] [Indexed: 12/02/2022] Open
Abstract
Purpose Epithelial–mesenchymal (EMT) transition plays an important role in metastasis and is accompanied by an upregulation of N-cadherin expression. A new nanoparticulate system (SPION/CCh/N-cad) based on superparamagnetic iron oxide nanoparticles, stabilized with a cationic derivative of chitosan and surface-modified with anti-N-cadherin antibody, was synthetized for the effective capture of N-cadherin expressing circulating tumor cells (CTC). Methods The morphology, physicochemical, and magnetic properties of the system were evaluated using dynamic light scattering (DLS), fluorescence spectroscopy, Mössbauer spectroscopy, magnetometry, and fluorescence spectroscopy. Atomic force microscopy (AFM), confocal microscopy and flow cytometry were used to study the interaction of our nanoparticulate system with N-cadherin expressed in prostate cancer cell lines (PC-3 and DU 145). A purpose-built cuvette was used in the cancer cell capture experiments. Results The obtained nanoparticles were a spherical, stable colloid, and exhibited excellent magnetic properties. Biological experiments confirmed that the novel SPION/CCh/N-cad system interacts specifically with N-cadherin present on the cell surface. Preliminary studies on the magnetic capture of PC-3 cells using the obtained nanoparticles were successful. Incubation times as short as 1 minute were sufficient for the synthesized system to effectively bind to the PC-3 cells. Conclusion Results obtained for our system suggest a possibility of using it to capture CTC in the flow conditions.
Collapse
Affiliation(s)
- Karolina Karnas
- Department of Chemistry, Jagiellonian University, Kraków, Poland.,Chair of Medical Biochemistry, Jagiellonian University Medical College, Kraków, Poland
| | - Tomasz Strączek
- AGH University of Science and Technology, Faculty of Physics and Applied Computer Science, Department of Solid State Physics, Kraków, Poland
| | - Czesław Kapusta
- AGH University of Science and Technology, Faculty of Physics and Applied Computer Science, Department of Solid State Physics, Kraków, Poland
| | - Małgorzata Lekka
- Department of Biophysical Microstructures, Institute of Nuclear Physics, Polish Academy of Sciences, Kraków, Poland
| | | | - Anna Karewicz
- Department of Chemistry, Jagiellonian University, Kraków, Poland
| |
Collapse
|
12
|
Quan Y, Zhang X, Butler W, Du Z, Wang M, Liu Y, Ping H. The role of N-cadherin/c-Jun/NDRG1 axis in the progression of prostate cancer. Int J Biol Sci 2021; 17:3288-3304. [PMID: 34512147 PMCID: PMC8416735 DOI: 10.7150/ijbs.63300] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Accepted: 07/06/2021] [Indexed: 02/03/2023] Open
Abstract
The dysregulation of androgen receptor (AR) signaling is a critical event in the progression of prostate cancer (PCa) and hormone therapy consisting of androgen deprivation (ADT) or AR inhibition is therefore used to treat advanced cases. It is known that N-cadherin becomes upregulated following ADT and can directly induce PCa transformation to the castration-resistant stage (CRPC). However, the relationship between AR and N-cadherin is unclear and may promote better understanding of CRPC pathogenesis and progression. Here, we demonstrate a new axis of N-cadherin/c-Jun/N-myc downstream regulated gene 1 (NDRG1) that N-cadherin promotes c-Jun expression and suppresses NDRG1 to promote invasion and migration of PCa cells through epithelial to mesenchymal transition (EMT). Targeting N-cadherin in combination with enzalutamide (ENZ) treatment synergistically suppressed PC3 cell proliferation in vivo and in vitro. Further studies showed that compared to lower Gleason score (GS) (GS < 7) cases, high GS (GS > 7) cases exhibited elevated N-cadherin expression and reduced NDRG1 expression, corroborating our in vitro observations. We further demonstrate that c-Jun, AR, and DNA methyltransferase-1 (DNMT1) form a complex in the 12-O-tetradecanoyl phorbol-13-acetate (TPA) response elements (TREs) region of the NDRG1 promoter, which suppresses NDRG1 transcription through DNA hypermethylation. In conclusion, we demonstrate an underlying mechanism for how N-cadherin collaborates with AR and NDRG1 to promote CRPC progression. Controlling N-cadherin/c-Jun/NDRG1 axis may help to overcome resistance to commonly used hormone therapy to improve long-term patient outcomes.
Collapse
Affiliation(s)
- Yongjun Quan
- Department of Urology, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China
| | - Xiaodong Zhang
- Department of Urology, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China
| | - William Butler
- Department of Pathology, Duke University School of Medicine, Durham NC 27710, USA
| | - Zhen Du
- Department of Urology, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China
| | - Mingdong Wang
- Department of Urology, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China
| | - Yuexin Liu
- Department of Urology, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China
| | - Hao Ping
- Department of Urology, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China
| |
Collapse
|
13
|
Zhao Y, Gao J, Xie X, Nan P, Liu F, Sun Y, Zhao X. BACH1 promotes the progression of esophageal squamous cell carcinoma by inducing the epithelial-mesenchymal transition and angiogenesis. Cancer Med 2021; 10:3413-3426. [PMID: 33932125 PMCID: PMC8124123 DOI: 10.1002/cam4.3884] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 03/11/2021] [Accepted: 03/13/2021] [Indexed: 02/06/2023] Open
Abstract
Metastasis to regional lymph nodes or distal organs predicts the progression of the disease and poor prognosis in esophageal squamous cell carcinoma (ESCC). Previous studies demonstrated that BTB and CNC homology 1 (BACH1) participates in various types of tumor metastasis. However, the function of BACH1 in ESCC was rarely reported. The present study demonstrated that BACH1 protein was overexpressed in ESCC tissues compared with paired esophageal epithelial tissues according to immunohistochemical staining (IHC). Higher levels of BACH1 mRNA were associated with decreased overall survival (OS) and shorter disease‐free survival (DFS) of ESCC patients based on an analysis of The Cancer Genome Atlas (TCGA) datasets. BACH1 significantly enhanced the migration and invasion of ESCC in vitro. Mechanistically, BACH1 promoted the epithelial–mesenchymal transition (EMT) by directly activating the transcription of CDH2, SNAI2, and VIM, as determined by chromatin immunoprecipitation‐quantitative polymerase chain reaction (ChIP‐qPCR). BACH1 overexpression significantly enhanced CDH2 promoter activity according to the results of a luciferase assay. The results of subsequent experiments indicated that BACH1 enhanced the growth of tumor xenografts. The density of CD31+ blood vessels and the expression of vascular endothelial growth factor C (VEGFC) in tumor xenografts were significantly associated with BACH1 levels according to the results of IHC and immunofluorescence (IF) analyses performed in vivo. Moreover, ChIP‐qPCR analysis demonstrated that the transcriptional activity of VEGFC was also upregulated by BACH1. Thus, BACH1 contributes to ESCC metastasis and tumorigenesis by partially facilitating the EMT and angiogenesis, and BACH1 may be a promising therapeutic target or molecular marker in ESCC.
Collapse
Affiliation(s)
- Yan Zhao
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jiajia Gao
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiufeng Xie
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Peng Nan
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Fang Liu
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yulin Sun
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiaohang Zhao
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
14
|
Namba N, Chonan Y, Nunokawa T, Sampetrean O, Saya H, Sudo R. Heterogeneous Glioma Cell Invasion Under Interstitial Flow Depending on Their Differentiation Status. Tissue Eng Part A 2021; 27:467-478. [PMID: 33403936 DOI: 10.1089/ten.tea.2020.0280] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Glioblastoma (GBM) is the most common and lethal type of malignant brain tumor. A deeper mechanistic understanding of the invasion of heterogeneous GBM cell populations is crucial to develop therapeutic strategies. A key regulator of GBM cell invasion is interstitial flow. However, the effect of an interstitial flow on the invasion of heterogeneous GBM cell populations composed of glioma initiating cells (GICs) and relatively differentiated progeny cells remains unclear. In the present study, we investigated how GICs invade three-dimensional (3D) hydrogels in response to an interstitial flow with respect to their differentiation status. Microfluidic culture systems were used to apply an interstitial flow to the cells migrating from the cell aggregates into the 3D hydrogel. Phase-contrast microscopy revealed that the invasion and protrusion formation of the GICs in differentiated cell conditions were significantly enhanced by a forward interstitial flow, whose direction was the same as that of the cell invasion, whereas those in stem cell conditions were not enhanced by the interstitial flow. The mechanism of flow-induced invasion was further investigated by focusing on differentiated cell conditions. Immunofluorescence images revealed that the expression of cell-extracellular matrix adhesion-associated molecules, such as integrin β1, focal adhesion kinase, and phosphorylated Src, was upregulated in forward interstitial flow conditions. We then confirmed that cell invasion and protrusion formation were significantly inhibited by PP2, a Src inhibitor. Finally, we observed that the flow-induced cell invasion was preceded by nestin-positive immature GICs at the invasion front and followed by tubulin β3-positive differentiated cells. Our findings provide insights into the development of novel therapeutic strategies to inhibit flow-induced glioma invasion. Impact statement A mechanistic understanding of heterogeneous glioblastoma cell invasion is crucial for developing therapeutic strategies. We observed that the invasion and protrusion formation of glioma initiating cells (GICs) were significantly enhanced by forward interstitial flow in differentiated cell conditions. The expression of integrin β1, focal adhesion kinase, and phosphorylated Src was upregulated, and the flow-induced invasion was significantly inhibited by a Src inhibitor. The flow-induced heterogeneous cell invasion was preceded by nestin-positive GICs at the invasion front and followed by tubulin β3-positive differentiated cells. Our findings provide insights into the development of novel therapeutic strategies to inhibit flow-induced glioma invasion.
Collapse
Affiliation(s)
- Naoko Namba
- Department of System Design Engineering, Keio University, Yokohama, Japan
| | - Yuta Chonan
- Department of System Design Engineering, Keio University, Yokohama, Japan
| | - Takehito Nunokawa
- Department of System Design Engineering, Keio University, Yokohama, Japan
| | - Oltea Sampetrean
- Division of Gene Regulation, Institute for Advanced Medical Research, Keio University School of Medicine, Tokyo, Japan
| | - Hideyuki Saya
- Division of Gene Regulation, Institute for Advanced Medical Research, Keio University School of Medicine, Tokyo, Japan
| | - Ryo Sudo
- Department of System Design Engineering, Keio University, Yokohama, Japan
| |
Collapse
|
15
|
Saxena K, Jolly MK, Balamurugan K. Hypoxia, partial EMT and collective migration: Emerging culprits in metastasis. Transl Oncol 2020; 13:100845. [PMID: 32781367 PMCID: PMC7419667 DOI: 10.1016/j.tranon.2020.100845] [Citation(s) in RCA: 131] [Impact Index Per Article: 26.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Revised: 07/12/2020] [Accepted: 07/27/2020] [Indexed: 02/07/2023] Open
Abstract
Epithelial-mesenchymal transition (EMT) is a cellular biological process involved in migration of primary cancer cells to secondary sites facilitating metastasis. Besides, EMT also confers properties such as stemness, drug resistance and immune evasion which can aid a successful colonization at the distant site. EMT is not a binary process; recent evidence suggests that cells in partial EMT or hybrid E/M phenotype(s) can have enhanced stemness and drug resistance as compared to those undergoing a complete EMT. Moreover, partial EMT enables collective migration of cells as clusters of circulating tumor cells or emboli, further endorsing that cells in hybrid E/M phenotypes may be the 'fittest' for metastasis. Here, we review mechanisms and implications of hybrid E/M phenotypes, including their reported association with hypoxia. Hypoxia-driven activation of HIF-1α can drive EMT. In addition, cyclic hypoxia, as compared to acute or chronic hypoxia, shows the highest levels of active HIF-1α and can augment cancer aggressiveness to a greater extent, including enriching for a partial EMT phenotype. We also discuss how metastasis is influenced by hypoxia, partial EMT and collective cell migration, and call for a better understanding of interconnections among these mechanisms. We discuss the known regulators of hypoxia, hybrid EMT and collective cell migration and highlight the gaps which needs to be filled for connecting these three axes which will increase our understanding of dynamics of metastasis and help control it more effectively.
Collapse
Affiliation(s)
- Kritika Saxena
- Centre for BioSystems Science and Engineering, Indian Institute of Science, Bangalore 560012, India
| | - Mohit Kumar Jolly
- Centre for BioSystems Science and Engineering, Indian Institute of Science, Bangalore 560012, India.
| | - Kuppusamy Balamurugan
- Laboratory of Cell and Developmental Signaling, Center for Cancer Research, National Cancer Institute, Frederick, MD 21702, USA.
| |
Collapse
|
16
|
Dong H, Sun S, Yan T, Liang C, Zhu J, Miao C, Qin C, Shao P, Wang Z, Li J, Li P. MicroRNA-195 inhibits proliferation and metastasis in renal cell carcinoma via regulating HMGA1. Am J Transl Res 2020; 12:2781-2792. [PMID: 32655809 PMCID: PMC7344069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Accepted: 04/29/2020] [Indexed: 06/11/2023]
Abstract
Growing evidence indicates that aberrant expression of microRNAs (miRNAs) contributes to tumorigenesis in various human malignancies. In this study we revealed that miR-195 acted as a tumor suppressor in renal cell carcinoma (RCC) through inhibition of HMGA1 expression. qRT-PCR was used to detect the miR-195 expression in RCC tissues and cell lines. RCC cell line Caki-1 and Caki-2 cells were used in this study. The luciferase report assay and rescue assay were performed to identify HMGA1 as the target gene of miR-195. Additionally, Kaplan-Meier method and log-rank test was used to explore the relationship between HMGA1 expression and RCC prognosis. We observed that miR-195 expression was significantly downregulated both in RCC tissues and in RCC cell lines. We observed that miR-195 overexpression inhibits the abilities of RCC cell proliferation, cell cycle progression and metastasis in vitro by targeting HMGA1 via epithelial to mesenchymal transition (EMT) pathway. In clinical specimens, HMGA1 was overexpressed in high-grade RCC when compared with its levels in normal tissues and low-grade RCC cancer, its expression levels were inversely correlated with overall survival. Our findings highlight an important role of miR-195 and HMGA1 in the molecular etiology of RCC, indicating that they can serve as potential biomarkers and therapy targets of RCC.
Collapse
Affiliation(s)
- Huiyu Dong
- State Key Laboratory of Reproductive Medicine and Department of Urology, The First Affiliated Hospital of Nanjing Medical UniversityNanjing, Jiangsu, China
| | - Shengjie Sun
- State Key Laboratory of Reproductive Medicine and Department of Urology, The First Affiliated Hospital of Nanjing Medical UniversityNanjing, Jiangsu, China
| | - Tao Yan
- State Key Laboratory of Reproductive Medicine and Department of Urology, The First Affiliated Hospital of Nanjing Medical UniversityNanjing, Jiangsu, China
| | - Chao Liang
- State Key Laboratory of Reproductive Medicine and Department of Urology, The First Affiliated Hospital of Nanjing Medical UniversityNanjing, Jiangsu, China
| | - Jundong Zhu
- Department of Urology, The First People’s Hospital of ChangzhouChangzhou, Jiangsu, China
| | - Chenkui Miao
- State Key Laboratory of Reproductive Medicine and Department of Urology, The First Affiliated Hospital of Nanjing Medical UniversityNanjing, Jiangsu, China
| | - Chao Qin
- State Key Laboratory of Reproductive Medicine and Department of Urology, The First Affiliated Hospital of Nanjing Medical UniversityNanjing, Jiangsu, China
| | - Pengfei Shao
- State Key Laboratory of Reproductive Medicine and Department of Urology, The First Affiliated Hospital of Nanjing Medical UniversityNanjing, Jiangsu, China
| | - Zengjun Wang
- State Key Laboratory of Reproductive Medicine and Department of Urology, The First Affiliated Hospital of Nanjing Medical UniversityNanjing, Jiangsu, China
| | - Jie Li
- State Key Laboratory of Reproductive Medicine and Department of Urology, The First Affiliated Hospital of Nanjing Medical UniversityNanjing, Jiangsu, China
| | - Pu Li
- State Key Laboratory of Reproductive Medicine and Department of Urology, The First Affiliated Hospital of Nanjing Medical UniversityNanjing, Jiangsu, China
| |
Collapse
|
17
|
Han P, Liu J, Lei Y, Lin Z, Tian D, Yan W. Netrin-1 promotes the collective cell migration of liver cancer cells in a 3D cell culture model. J Physiol Biochem 2019; 75:489-498. [PMID: 31407237 DOI: 10.1007/s13105-019-00701-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Accepted: 08/04/2019] [Indexed: 01/01/2023]
Abstract
Collective cell migration plays an important role in embryonic development, wound healing, and cancer metastasis. We aimed to investigate the expression, role, and mechanism of Netrin-1 in collective cell migration using a3D culture model. An immunohistochemical study showed that certain cells invaded surrounding tissue by collective migration and that Netrin-1 expression in these cells was increased, especially at the invasive front. In the 3D culture model, collective cell migration was clearly observed, as leader cells were followed by cells migrating along a canal. N-cadherin-mediated cell junctions were observed in collective cell migration, and Netrin-1 expression was elevated in these cells. Netrin-1 did not affect the expression of N-cadherin in 2D-cultured cells; however, in 3D culture, the overexpression of Netrin-1 increased N-cadherin and promoted the collective migration of Huh7 cells, while the knockdown of Netrin-1 decreased N-cadherin and inhibited collective migration in SK-Hep-1 cells. Interestingly, N-cadherin knockdown in Huh7 cells significantly diminished Netrin-1-promoted collective cell migration, while the overexpression of N-cadherin restored collective migration in Netrin-1-knockdown SK-Hep1 cells. These results suggest that Netrin-1 enhances N-cadherin junctions to promote liver cancer cell collective migration in 3D cell culture and may subsequently increase liver cancer metastasis.
Collapse
Affiliation(s)
- Ping Han
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095# Jiefang Avenue, Wuhan, 430030, People's Republic of China
| | - Jingmei Liu
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095# Jiefang Avenue, Wuhan, 430030, People's Republic of China
| | - Yu Lei
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095# Jiefang Avenue, Wuhan, 430030, People's Republic of China
| | - Zhuoying Lin
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095# Jiefang Avenue, Wuhan, 430030, People's Republic of China
| | - Dean Tian
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095# Jiefang Avenue, Wuhan, 430030, People's Republic of China.
| | - Wei Yan
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095# Jiefang Avenue, Wuhan, 430030, People's Republic of China.
| |
Collapse
|
18
|
Wang X, Enomoto A, Weng L, Mizutani Y, Abudureyimu S, Esaki N, Tsuyuki Y, Chen C, Mii S, Asai N, Haga H, Ishida S, Yokota K, Akiyama M, Takahashi M. Girdin/GIV regulates collective cancer cell migration by controlling cell adhesion and cytoskeletal organization. Cancer Sci 2018; 109:3643-3656. [PMID: 30194792 PMCID: PMC6215880 DOI: 10.1111/cas.13795] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Revised: 09/03/2018] [Accepted: 09/05/2018] [Indexed: 12/28/2022] Open
Abstract
Pathological observations show that cancer cells frequently invade the surrounding stroma in collective groups rather than through single cell migration. Here, we studied the role of the actin-binding protein Girdin, a specific regulator of collective migration of neuroblasts in the brain, in collective cancer cell migration. We found that Girdin was essential for the collective migration of the skin cancer cell line A431 on collagen gels as well as their fibroblast-led collective invasion in an organotypic culture model. We provide evidence that Girdin binds to β-catenin that plays important roles in the Wnt signaling pathway and in E-cadherin-mediated cell-cell adhesion. Girdin-depleted cells displayed scattering and impaired E-cadherin-specific cell-cell adhesion. Importantly, Girdin depletion led to impaired cytoskeletal association of the β-catenin complex, which was accompanied by changes in the supracellular actin cytoskeletal organization of cancer cell cohorts on collagen gels. Although the underlying mechanism is unclear, this observation is consistent with the established role of the actin cytoskeletal system and cell-cell adhesion in the collective behavior of cells. Finally, we showed the correlation of the expression of Girdin with that of the components of the E-cadherin complex and the differentiation of human skin cancer. Collectively, our results suggest that Girdin is an important modulator of the collective behavior of cancer cells.
Collapse
Affiliation(s)
- Xiaoze Wang
- Department of PathologyNagoya University Graduate School of MedicineNagoyaJapan
| | - Atsushi Enomoto
- Department of PathologyNagoya University Graduate School of MedicineNagoyaJapan
| | - Liang Weng
- Department of PathologyNagoya University Graduate School of MedicineNagoyaJapan
| | - Yasuyuki Mizutani
- Department of PathologyNagoya University Graduate School of MedicineNagoyaJapan
| | - Shaniya Abudureyimu
- Department of PathologyNagoya University Graduate School of MedicineNagoyaJapan
| | - Nobutoshi Esaki
- Department of PathologyNagoya University Graduate School of MedicineNagoyaJapan
| | - Yuta Tsuyuki
- Department of PathologyNagoya University Graduate School of MedicineNagoyaJapan
| | - Chen Chen
- Department of PathologyNagoya University Graduate School of MedicineNagoyaJapan
| | - Shinji Mii
- Department of PathologyNagoya University Graduate School of MedicineNagoyaJapan
| | - Naoya Asai
- Department of PathologyNagoya University Graduate School of MedicineNagoyaJapan
| | - Hisashi Haga
- Transdisciplinary Life Science CourseFaculty of Advanced Life ScienceHokkaido UniversitySapporoJapan
| | - Sumire Ishida
- Transdisciplinary Life Science CourseFaculty of Advanced Life ScienceHokkaido UniversitySapporoJapan
| | - Kenji Yokota
- Department of DermatologyNagoya University Graduate School of MedicineNagoyaJapan
| | - Masashi Akiyama
- Department of DermatologyNagoya University Graduate School of MedicineNagoyaJapan
| | - Masahide Takahashi
- Department of PathologyNagoya University Graduate School of MedicineNagoyaJapan
| |
Collapse
|
19
|
Hua Y, Liang C, Miao C, Wang S, Su S, Shao P, Liu B, Bao M, Zhu J, Xu A, Zhang J, Li J, Wang Z. MicroRNA-126 inhibits proliferation and metastasis in prostate cancer via regulation of ADAM9. Oncol Lett 2018; 15:9051-9060. [PMID: 29805636 PMCID: PMC5958673 DOI: 10.3892/ol.2018.8528] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Accepted: 02/27/2018] [Indexed: 01/01/2023] Open
Abstract
The aberrant expression of microRNAs (miRs) has been identified to serve a crucial role in tumor progression. The present study aimed to evaluate the role of miR-126 in human prostate cancer (PCa). Firstly, miR-126 expression in prostate cancer tissues and cell lines was analyzed. A luciferase reporter assay and a rescue assay were performed, which identified ADAM metalloproteinase domain 9 (ADAM9) as the target gene of miR-126. Subsequently, Kaplan-Meier and log-rank analyses were used to investigate the association between ADAM9 expression and PCa prognosis. The results revealed that miR-126 expression was significantly downregulated in PCa tissues and cell lines. miR-126 overexpression was demonstrated to reduce PCa cell proliferation and metastasis, and to reverse the epithelial-mesenchymal transition process in vitro. In addition, as the target gene of miR-126, the upregulation of ADAM9 reestablished cell functions, including cell proliferation, migration and invasion. Patients with high ADAM9 expression levels exhibited a shorter biochemical recurrence-free survival time. In summary, miR-126 serves a role in the proliferation and metastasis of PCa cells, indicating that miR-126 and ADAM9 may represent potential biomarkers in the progression of advanced PCa, in addition to therapeutic targets.
Collapse
Affiliation(s)
- Yibo Hua
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Chao Liang
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Chenkui Miao
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Shangqian Wang
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Shifeng Su
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Pengfei Shao
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Bianjiang Liu
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Meiling Bao
- Department of Pathology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Jundong Zhu
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Aiming Xu
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Jianzhong Zhang
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Jie Li
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Zengjun Wang
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| |
Collapse
|
20
|
Ciołczyk-Wierzbicka D, Laidler P. The inhibition of invasion of human melanoma cells through N-cadherin knock-down. Med Oncol 2018; 35:42. [PMID: 29492694 PMCID: PMC5830464 DOI: 10.1007/s12032-018-1104-9] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Accepted: 02/23/2018] [Indexed: 12/27/2022]
Abstract
N-cadherin seems to promote cell migration and invasion in many types of cancers. The object of this study is recognition of the possible role of N-cadherin and selected downstream protein kinases: PI3K, ERK1/2, and mTOR in cell invasion in malignant melanoma. Melanoma cells were transfected with the small interfering RNA (siRNA) that targets human N-cadherin gene (CDH2). Inhibitors LY294002 (PI3K), U0126 (ERK1/2), and everolimus (mTOR) were used to inhibit selected kinases of signalling pathways. In vitro cell invasion was studied using Matrigel and an analysis of matrix metalloproteinases MMP-2 and MMP-9 activity by gelatinase zymogram assay. Treatment of melanoma cell with either siRNA against N-cadherin or protein kinase inhibitors led to significantly decreased MMPs expression and activity, as well as diminished invasion. Both the current and the former results suggest that activation of PI3/AKT, mTOR, and ERK kinase, following N-cadherin expression, contributes not only to increased proliferation but also invasive potential of melanoma cells. The results also indicate that N-cadherin, as well as the studied kinases, should be considered as a potential target in melanoma therapy.
Collapse
Affiliation(s)
- Dorota Ciołczyk-Wierzbicka
- Chair of Medical Biochemistry, Jagiellonian University Medical College, ul. Kopernika 7, 31-034, Kraków, Poland.
| | - Piotr Laidler
- Chair of Medical Biochemistry, Jagiellonian University Medical College, ul. Kopernika 7, 31-034, Kraków, Poland
| |
Collapse
|
21
|
Silva RDS, Lombardi APG, de Souza DS, Vicente CM, Porto CS. Activation of estrogen receptor beta (ERβ) regulates the expression of N-cadherin, E-cadherin and β-catenin in androgen-independent prostate cancer cells. Int J Biochem Cell Biol 2018; 96:40-50. [PMID: 29341930 DOI: 10.1016/j.biocel.2018.01.008] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Revised: 01/10/2018] [Accepted: 01/12/2018] [Indexed: 12/11/2022]
Abstract
The aim of the present study was to investigate the impact of the activation of estrogen receptors on expression and localization of N-cadherin, E-cadherin and non-phosphorylated β-catenin in androgen-independent prostate cancer cells (PC-3 and DU-145) and in human post pubertal prostate epithelial cells (PNT1A). Expression of N-cadherin was detected in PNT1A and PC-3 cells, but not in DU-145 cells. E-cadherin was detected only in DU-145 cells and β-catenin was detected in all cells studied. N-cadherin and β-catenin were located preferentially in the cellular membrane of PNT1A cells and in the cytoplasm of PC-3 cells. E-cadherin and β-catenin were located preferentially in the cellular membrane of DU-145 cells. 17β-estradiol (E2) or the ERα-selective agonist PPT did not affect the content and localization of N-cadherin in PC-3 and PNT1A cells or E-cadherin in DU-145 cells. In PC-3 cells, ERβ-selective agonist DPN decreased the expression of N-cadherin. DPN-induced downregulation of N-cadherin was blocked by pretreatment with the ERβ-selective antagonist (PHTPP), indicating that ERβ1 is the upstream receptor regulating the expression of N-cadherin. In DU-145 cells, the activation of ERβ1 by DPN increased the expression of E-cadherin. Taken together, these results suggest that activation of ERβ1 is required to maintain an epithelial phenotype in PC-3 and DU-145 cells. The activation of ERβ1 also increased the expression of β-catenin in cytoplasm of PC-3 and in the cellular membrane of DU-145 cells. In conclusion, our results indicate differential expression and localization of N-cadherin, E-cadherin and β-catenin in androgen-independent prostate cancer cells. The reduction of N-cadherin content by activation of ERβ, exclusively observed in androgen-independent prostate cancer cells (PC-3), may be related to the activation of signaling pathways, such as the release of β-catenin into the cytoplasm, translocation of β-catenin to the nucleus and activation of gene transcription.
Collapse
Affiliation(s)
- Rafael de Souza Silva
- Section of Experimental Endocrinology, Department of Pharmacology, Escola Paulista de Medicina, Universidade Federal de São Paulo, Rua Pedro de Toledo 669, Vila Clementino, São Paulo, SP, 04039-032, Brazil
| | - Ana Paola G Lombardi
- Section of Experimental Endocrinology, Department of Pharmacology, Escola Paulista de Medicina, Universidade Federal de São Paulo, Rua Pedro de Toledo 669, Vila Clementino, São Paulo, SP, 04039-032, Brazil
| | - Deborah Simão de Souza
- Section of Experimental Endocrinology, Department of Pharmacology, Escola Paulista de Medicina, Universidade Federal de São Paulo, Rua Pedro de Toledo 669, Vila Clementino, São Paulo, SP, 04039-032, Brazil
| | - Carolina M Vicente
- Section of Experimental Endocrinology, Department of Pharmacology, Escola Paulista de Medicina, Universidade Federal de São Paulo, Rua Pedro de Toledo 669, Vila Clementino, São Paulo, SP, 04039-032, Brazil
| | - Catarina S Porto
- Section of Experimental Endocrinology, Department of Pharmacology, Escola Paulista de Medicina, Universidade Federal de São Paulo, Rua Pedro de Toledo 669, Vila Clementino, São Paulo, SP, 04039-032, Brazil.
| |
Collapse
|
22
|
Dasen B, Vlajnic T, Mengus C, Ruiz C, Bubendorf L, Spagnoli G, Wyler S, Erne P, Resink TJ, Philippova M. T-cadherin in prostate cancer: relationship with cancer progression, differentiation and drug resistance. JOURNAL OF PATHOLOGY CLINICAL RESEARCH 2016; 3:44-57. [PMID: 28138401 PMCID: PMC5259566 DOI: 10.1002/cjp2.61] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Revised: 09/30/2016] [Accepted: 10/16/2016] [Indexed: 01/17/2023]
Abstract
Prostate cancer represents the second leading cause of cancer-related death in men. T-cadherin (CDH13) is an atypical GPI-anchored member of the cadherin family of adhesion molecules. Its gene was reported to be downregulated in a small series of prostate tumours. T-cadherin protein expression/localisation in prostate tissue has never been investigated. The purpose of our study was to analyse CDH13 gene and protein levels in large sets of healthy and cancer prostate tissue specimens and evaluate CDH13 effects on the sensitivity of prostate cancer cells to chemotherapy. Analysis of CDH13 gene expression in the TCGA RNAseq dataset for prostate adenocarcinoma (N = 550) and in tissue samples (N = 101) by qPCR revealed weak positive correlation with the Gleason score in cancer and no difference between benign and malignant specimens. Immunohistochemical analysis of tissue sections (N = 12) and microarrays (N = 128 specimens) demonstrated the presence of CDH13 on the apical surface and at intercellular contacts of cytokeratin 8-positive luminal cells and cells double-positive for cytokeratin 8 and basal marker p63. T-cadherin protein expression was markedly upregulated in cancer as compared to benign prostate hyperplasia, the increase being more prominent in organ-confined than in advanced hormone-resistant tumours, and correlated negatively with the Gleason pattern. T-cadherin protein level correlated strongly with cytokeratin 8 and with an abnormal diffuse/membrane localisation pattern of p63. Ectopic expression of CDH13 in metastatic prostate cancer cell line DU145 reduced cell growth in the presence of doxorubicin. We conclude that CDH13 protein, but not its gene expression, is strongly upregulated in early prostate cancer, correlates with changes in luminal/basal differentiation and p63 localisation, and promotes sensitivity of cancer cells to doxorubicin. These data identify CDH13 as a novel molecule relevant for prostate cancer progression and response to therapy.
Collapse
Affiliation(s)
- Boris Dasen
- Department of Biomedicine, Laboratory for Signal Transduction University Hospital Basel Switzerland
| | - Tatjana Vlajnic
- Institute of Pathology, University Hospital Basel Switzerland
| | - Chantal Mengus
- Institute of Surgical Research and Department of Biomedicine Basel University Hospital Switzerland
| | - Christian Ruiz
- Institute of Pathology, University Hospital Basel Switzerland
| | - Lukas Bubendorf
- Institute of Pathology, University Hospital Basel Switzerland
| | - Giulio Spagnoli
- Institute of Surgical Research and Department of Biomedicine Basel University Hospital Switzerland
| | - Stephen Wyler
- Urology Clinic, University Hospital Basel Switzerland
| | - Paul Erne
- Department of Biomedicine, Laboratory for Signal Transduction University Hospital Basel Switzerland
| | - Thérèse J Resink
- Department of Biomedicine, Laboratory for Signal Transduction University Hospital Basel Switzerland
| | - Maria Philippova
- Department of Biomedicine, Laboratory for Signal Transduction University Hospital Basel Switzerland
| |
Collapse
|
23
|
Przybyla L, Muncie JM, Weaver VM. Mechanical Control of Epithelial-to-Mesenchymal Transitions in Development and Cancer. Annu Rev Cell Dev Biol 2016; 32:527-554. [DOI: 10.1146/annurev-cellbio-111315-125150] [Citation(s) in RCA: 93] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Laralynne Przybyla
- Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California, San Francisco, California 94143;
| | - Jonathon M. Muncie
- Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California, San Francisco, California 94143;
- Joint Graduate Group in Bioengineering (University of California, San Francisco, and University of California, Berkeley), San Francisco, California 94143
| | - Valerie M. Weaver
- Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California, San Francisco, California 94143;
- Departments of Anatomy, Bioengineering, and Therapeutic Sciences, Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, The Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California 94143
| |
Collapse
|
24
|
Yang C, Li X, Wang C, Fu S, Li H, Guo Z, Zhao S, Lin J. N-cadherin regulates beta-catenin signal and its misexpression perturbs commissural axon projection in the developing chicken spinal cord. J Mol Histol 2016; 47:541-554. [PMID: 27650519 DOI: 10.1007/s10735-016-9698-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Accepted: 09/14/2016] [Indexed: 12/31/2022]
Abstract
N-cadherin is a calcium-sensitive cell adhesion molecule that plays an important role in the formation of the neural circuit and the development of the nervous system. In the present study, we investigated the function of N-cadherin in cell-cell connection in vitro with HEK293T cells, and in commissural axon projections in the developing chicken spinal cord using in ovo electroporation. Cell-cell connections increased with N-cadherin overexpression in HEK293T cells, while cell contacts disappeared after co-transfection with an N-cadherin-shRNA plasmid. The knockdown of N-cadherin caused the accumulation of β-catenin in the nucleus, supporting the notion that N-cadherin regulates β-catenin signaling in vitro. Furthermore, N-cadherin misexpression perturbed commissural axon projections in the spinal cord. The overexpression of N-cadherin reduced the number of axons that projected alongside the contralateral margin of the floor plate, and formed intermediate longitudinal commissural axons. In contrast, the knockdown of N-cadherin perturbed commissural axon projections significantly, affecting the projections alongside the contralateral margin of the floor plate, but did not affect intermediate longitudinal commissural axons. Taken together, these findings suggest that N-cadherin regulates commissural axon projections in the developing chicken spinal cord.
Collapse
Affiliation(s)
- Ciqing Yang
- College of Life Science and Technology, Xinxiang Medical University, Xinxiang, 453003, China.,Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang, 453003, China
| | - Xiaoying Li
- College of Life Science and Technology, Xinxiang Medical University, Xinxiang, 453003, China
| | - Congrui Wang
- Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang, 453003, China
| | - Sulei Fu
- College of Life Science and Technology, Xinxiang Medical University, Xinxiang, 453003, China
| | - Han Li
- College of Life Science and Technology, Xinxiang Medical University, Xinxiang, 453003, China.,Advanced Medical and Dental Institute, Universiti Sains Malaysia, 13200, Bertam, Penang, Malaysia
| | - Zhikun Guo
- Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang, 453003, China
| | - Shanting Zhao
- College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, China
| | - Juntang Lin
- College of Life Science and Technology, Xinxiang Medical University, Xinxiang, 453003, China. .,Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang, 453003, China. .,Institute of Anatomy I, Jena University Hospital, 07743, Jena, Germany.
| |
Collapse
|
25
|
Cheng G, Wang S, Li X, Li S, Zheng Y, Zhang L, Bao M, Liang C, Huang Z, Liu Y, Qin C, Shao P, Li J, Hua L, Yin C, Wang Z. Positive expression of NR6A1/CT150 as a predictor of biochemical recurrence-free survival in prostate cancer patients. Oncotarget 2016; 8:64427-64439. [PMID: 28969082 PMCID: PMC5610014 DOI: 10.18632/oncotarget.11749] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2015] [Accepted: 07/26/2016] [Indexed: 01/15/2023] Open
Abstract
NR6A1/CT150, as an orphan receptor, is a novel member of the cancer-testis (CT) antigen family. Here, we investigated the expression and function of NR6A1 and its underlying mechanisms in prostate cancer (PCa) patients who underwent radical prostatectomy. A total of 303 cases of prostate cancer after radical prostatectomy were analysed in a tissue microarray (TMA) for NR6A1 immunohistochemistry-based protein expression. Kaplan–Meier/log-rank analysis and Cox regression analysis were used to investigate the relationship between NR6A1 expression and clinicopathological factors in PCa. NR6A1 mRNA expression was examined by reversing transcriptase-polymerase chain reaction (RT-PCR). Knockdown of NR6A1 by small interfering RNA mediated gene silencing and overexpression of NR6A1 through lentivirus were utilized to investigate its potential role in prostate cancer cells. NR6A1 protein expression was 29.7% (90/303) and mRNA expression was 28.1%(9/32) in PCa patients. NR6A1 expression was significantly associated with Gleason score (GS) (P=0.003) and tumor stage (P=0.042). The patients with positive NR6A1 expression have a shorter biochemical recurrence-free survival. NR6A1 predicted biochemical recurrence in univariate (P=0.0159) and multivariate models (P=0.0317). In addition, gene silencing of NR6A1 resulted in G0/G1 phase cell cycle arrest, and decreased metastatic and invasive potential of prostate cancer cells DU145 and PC3. In contrast, overexpression of NR6A1 reduced G0/G1 phase cell cycle arrest, and promoted metastatic and invasive potential of prostate cancer cells 22RV1. And overexpression of NR6A1 significantly promoted tumor growth in vivo. What's more, down regulation of NR6A1 could reverse epithelial-to-mesenchymal transition (EMT) process in DU145 and PC3 cell lines, and the overexpression could enhance EMT process in 22RV1 cell line. NR6A1 played a prominent role in migration and invasion of PCa cells, and it is indicated that NR6A1 may act as a novel marker for biochemical recurrence after radical prostatectomy.
Collapse
Affiliation(s)
- Gong Cheng
- Department of Urology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Shangqian Wang
- Department of Urology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xiao Li
- Department of Urology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China.,Department of Urology, the Affiliated Cancer Hospital of Jiangsu Province of Nanjing Medical University, Nanjing, China
| | - Shuang Li
- Department of Urology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yang Zheng
- Department of Urology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Lei Zhang
- Department of Urology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Meiling Bao
- Department of Urology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China.,Department of Pathology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Chao Liang
- Department of Urology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Zhengkai Huang
- Department of Urology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yiyang Liu
- Department of Urology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Chao Qin
- Department of Urology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Pengfei Shao
- Department of Urology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jie Li
- Department of Urology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Lixin Hua
- Department of Urology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Changjun Yin
- Department of Urology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Zengjun Wang
- Department of Urology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
26
|
Verrill C, Cerundolo L, Mckee C, White M, Kartsonaki C, Fryer E, Morris E, Brewster S, Ratnayaka I, Marsden L, Lilja H, Muschel R, Lu X, Hamdy F, Bryant RJ. Altered expression of epithelial-to-mesenchymal transition proteins in extraprostatic prostate cancer. Oncotarget 2016; 7:1107-19. [PMID: 26701730 PMCID: PMC4811447 DOI: 10.18632/oncotarget.6689] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Accepted: 12/05/2015] [Indexed: 01/09/2023] Open
Abstract
Epithelial to mesenchymal transition (EMT) of cancer cells involves loss of epithelial polarity and adhesiveness, and gain of invasive and migratory mesenchymal behaviours. EMT occurs in prostate cancer (PCa) but it is unknown whether this is in specific areas of primary tumours. We examined whether any of eleven EMT-related proteins have altered expression or subcellular localisation within the extraprostatic extension component of locally advanced PCa compared with other localisations, and whether similar changes may occur in in vitro organotypic PCa cell cultures and in vivo PCa models. Expression profiles of three proteins (E-cadherin, Snail, and α-smooth muscle actin) were significantly different in extraprostatic extension PCa compared with intra-prostatic tumour, and 18/27 cases had an expression change of at least one of these three proteins. Of the three significantly altered EMT proteins in pT3 samples, one showed similar significantly altered expression patterns in in vitro organotypic culture models, and two in in vivo Pten-/- model samples. These results suggest that changes in EMT protein expression can be observed in the extraprostatic extension component of locally invasive PCa. The biology of some of these changes in protein expression may be studied in certain in vitro and in vivo PCa models.
Collapse
Affiliation(s)
- Clare Verrill
- Department of Cellular Pathology, Oxford University Hospitals NHS Foundation Trust, John Radcliffe Hospital, Headington, Oxford, UK
- Nuffield Department of Surgical Sciences, University of Oxford, Headington, Oxford, UK
| | - Lucia Cerundolo
- Nuffield Department of Surgical Sciences, University of Oxford, Headington, Oxford, UK
| | - Chad Mckee
- CRUK/MRC Oxford Institute for Radiation Oncology, University of Oxford, Headington, Oxford, UK
| | - Michael White
- Ludwig Institute for Cancer Research Ltd, University of Oxford, Nuffield Department of Clinical Medicine, Headington, Oxford, UK
| | | | - Eve Fryer
- Department of Cellular Pathology, Oxford University Hospitals NHS Foundation Trust, John Radcliffe Hospital, Headington, Oxford, UK
| | - Emma Morris
- Nuffield Department of Surgical Sciences, University of Oxford, Headington, Oxford, UK
- Ludwig Institute for Cancer Research Ltd, University of Oxford, Nuffield Department of Clinical Medicine, Headington, Oxford, UK
| | - Simon Brewster
- Department of Urology, Churchill Hospital, Headington, Oxford, UK
| | - Indrika Ratnayaka
- Ludwig Institute for Cancer Research Ltd, University of Oxford, Nuffield Department of Clinical Medicine, Headington, Oxford, UK
| | - Luke Marsden
- Nuffield Department of Surgical Sciences, University of Oxford, Headington, Oxford, UK
| | - Hans Lilja
- Nuffield Department of Surgical Sciences, University of Oxford, Headington, Oxford, UK
- Departments of Surgery (Urology Service), Laboratory Medicine (Clinical Chemistry Service) and Medicine (Genitourinary Oncology Service), Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Laboratory Medicine and Clinical Sciences in Malmö, Lund University, Skåne University Hospital, Malmö, Sweden
- Institute of Biomedical Technology, University of Tampere, Tampere, Finland
| | - Ruth Muschel
- CRUK/MRC Oxford Institute for Radiation Oncology, University of Oxford, Headington, Oxford, UK
| | - Xin Lu
- Ludwig Institute for Cancer Research Ltd, University of Oxford, Nuffield Department of Clinical Medicine, Headington, Oxford, UK
| | - Freddie Hamdy
- Nuffield Department of Surgical Sciences, University of Oxford, Headington, Oxford, UK
| | - Richard J. Bryant
- Nuffield Department of Surgical Sciences, University of Oxford, Headington, Oxford, UK
- Ludwig Institute for Cancer Research Ltd, University of Oxford, Nuffield Department of Clinical Medicine, Headington, Oxford, UK
| |
Collapse
|
27
|
Puliafito A, De Simone A, Seano G, Gagliardi PA, Di Blasio L, Chianale F, Gamba A, Primo L, Celani A. Three-dimensional chemotaxis-driven aggregation of tumor cells. Sci Rep 2015; 5:15205. [PMID: 26471876 PMCID: PMC4607978 DOI: 10.1038/srep15205] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2015] [Accepted: 09/21/2015] [Indexed: 12/11/2022] Open
Abstract
One of the most important steps in tumor progression involves the transformation from a differentiated epithelial phenotype to an aggressive, highly motile phenotype, where tumor cells invade neighboring tissues. Invasion can occur either by isolated mesenchymal cells or by aggregates that migrate collectively and do not lose completely the epithelial phenotype. Here, we show that, in a three-dimensional cancer cell culture, collective migration of cells eventually leads to aggregation in large clusters. We present quantitative measurements of cluster velocity, coalescence rates, and proliferation rates. These results cannot be explained in terms of random aggregation. Instead, a model of chemotaxis-driven aggregation - mediated by a diffusible attractant - is able to capture several quantitative aspects of our results. Experimental assays of chemotaxis towards culture conditioned media confirm this hypothesis. Theoretical and numerical results further suggest an important role for chemotactic-driven aggregation in spreading and survival of tumor cells.
Collapse
Affiliation(s)
| | - Alessandro De Simone
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland
| | - Giorgio Seano
- Candiolo Cancer Institute FPO-IRCCS, Candiolo, Turin, Italy.,Edwin L. Steele Laboratory for Tumor Biology, Harvard Medical School, Massachusetts General Hospital, Boston, MA, USA
| | - Paolo Armando Gagliardi
- Candiolo Cancer Institute FPO-IRCCS, Candiolo, Turin, Italy.,Department of Oncology, University of Turin, Turin 10060, Italy
| | - Laura Di Blasio
- Candiolo Cancer Institute FPO-IRCCS, Candiolo, Turin, Italy.,Department of Oncology, University of Turin, Turin 10060, Italy
| | | | - Andrea Gamba
- Institute of Condensed Matter Physics and Complex Systems, Department of Applied Science and Technology, Polytechnic University of Turin, Corso Duca degli Abruzzi, 24, 10129 Torino, Italy.,Human Genetics Foundation (HuGeF), Via Nizza 52, Torino, Italy.,Istituto Nazionale di Fisica Nucleare (INFN), Torino, Via Giuria 1, 10125 Torino, Italy
| | - Luca Primo
- Candiolo Cancer Institute FPO-IRCCS, Candiolo, Turin, Italy.,Department of Oncology, University of Turin, Turin 10060, Italy
| | - Antonio Celani
- Quantitative Life Sciences Unit, The Abdus Salam Center for Theoretical Physics (ICTP), Strada Costiera 11, I-34151 Trieste, Italy
| |
Collapse
|
28
|
Gueron G, Giudice J, Valacco P, Paez A, Elguero B, Toscani M, Jaworski F, Leskow FC, Cotignola J, Marti M, Binaghi M, Navone N, Vazquez E. Heme-oxygenase-1 implications in cell morphology and the adhesive behavior of prostate cancer cells. Oncotarget 2015; 5:4087-102. [PMID: 24961479 PMCID: PMC4147308 DOI: 10.18632/oncotarget.1826] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Prostate cancer (PCa) is the second leading cause of cancer death in men. Although previous studies in PCa have focused on cell adherens junctions (AJs), key players in metastasis, they have left the molecular mechanisms unexplored. Inflammation and the involvement of reactive oxygen species (ROS) are critical in the regulation of cell adhesion and the integrity of the epithelium. Heme oxygenase-1 (HO-1) counteracts oxidative and inflammatory damage. Here, we investigated whether HO-1 is implicated in the adhesive and morphological properties of tumor cells. Genes differentially regulated by HO-1 were enriched for cell motility and adhesion biological processes. HO-1 induction, increased E-cadherin and β-catenin levels. Immunofluorescence analyses showed a striking remodeling of E-cadherin/β-catenin based AJs under HO-1 modulation. Interestingly, the enhanced levels of E-cadherin and β-catenin coincided with a markedly change in cell morphology. To further our analysis we sought to identify HO-1 binding proteins that might participate in the regulation of cell morphology. A proteomics approach identified Muskelin, as a novel HO-1 partner, strongly implicated in cell morphology regulation. These results define a novel role for HO-1 in modulating the architecture of cell-cell interactions, favoring a less aggressive phenotype and further supporting its anti-tumoral function in PCa.
Collapse
|
29
|
N-cadherin participated in invasion and metastasis of human esophageal squamous cell carcinoma via taking part in the formation of vasculogenic mimicry. Med Oncol 2015; 32:480. [PMID: 25575439 DOI: 10.1007/s12032-014-0480-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2014] [Accepted: 12/20/2014] [Indexed: 12/13/2022]
Abstract
Vasculogenic mimicry (VM) refers to the unique ability of highly aggressive tumor cells to mimic the pattern of embryonic vasculogenic networks, and the presence of VM correlates to an increased risk of metastasis and poor clinical outcome of cancers. Several key molecules, including N-cadherin, have been implicated in VM. However, the role of N-cadherin in the formation of VM in esophageal squamous cell carcinoma (ESCC) had not been elucidated. In this study, firstly we aimed to identify VM patterns in ESCC tissues and to explore their clinical significance. VM was present in 12 out of 56 samples, and ESCC with lymph node metastasis had a higher incidence of VM than that without lymph node metastasis. More importantly, VM channels were associated with the expression of N-cadherin in ESCC tissues. In order to further explore the role of N-cadherin in VM formation and invasion and metastasis in ESCC, secondly, we silenced the expression of N-cadherin with small hairpin RNA in ESCC cell line KYSE-70; herein, we showed that KYSE-70 cells with N-cadherin silencing lost not only the capacity to form tube-like structures on collagen (VM) but also the invasion, metastasis and proliferation ability in KYSE-70 cells in vitro. Taken together, antivascular therapies targeting tumor cell VM may be an effective approach to the treatment of patients with highly metastatic ESCC.
Collapse
|
30
|
Nestin depletion induces melanoma matrix metalloproteinases and invasion. J Transl Med 2014; 94:1382-95. [PMID: 25365206 PMCID: PMC4419570 DOI: 10.1038/labinvest.2014.130] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2014] [Revised: 07/25/2014] [Accepted: 09/08/2014] [Indexed: 12/20/2022] Open
Abstract
Matrix metalloproteinases (MMPs) are key biological mediators of processes as diverse as wound healing, embryogenesis, and cancer progression. Although MMPs may be induced through multiple signaling pathways, the precise mechanisms for their regulation in cancer are incompletely understood. Because cytoskeletal changes are known to accompany MMP expression, we sought to examine the potential role of the poorly understood cytoskeletal protein, nestin, in modulating melanoma MMPs. Nestin knockdown (KD) upregulated the expression of specific MMPs and MMP-dependent invasion both through extracellular matrix barriers in vitro and in peritumoral connective tissue of xenografts in vivo. The development of three-dimensional melanospheres that in vitro partially recapitulate noninvasive tumorigenic melanoma growth was inhibited by nestin KD, although ECM invasion by aberrant melanospheres that did form was enhanced. Mechanistically, nestin KD-dependent melanoma invasion was associated with intracellular redistribution of phosphorylated focal adhesion kinase and increased melanoma cell responsiveness to transforming growth factor-beta, both implicated in pathways of melanoma invasion. The results suggest that the heretofore poorly understood intermediate filament, nestin, may serve as a novel mediator of MMPs critical to melanoma virulence.
Collapse
|
31
|
Maslova K, Kyriakakis E, Pfaff D, Frachet A, Frismantiene A, Bubendorf L, Ruiz C, Vlajnic T, Erne P, Resink TJ, Philippova M. EGFR and IGF-1R in regulation of prostate cancer cell phenotype and polarity: opposing functions and modulation by T-cadherin. FASEB J 2014; 29:494-507. [PMID: 25381040 DOI: 10.1096/fj.14-249367] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
T-cadherin is an atypical glycosylphosphatidylinsoitol-anchored member of the cadherin superfamily of adhesion molecules. We found that T-cadherin overexpression in malignant (DU145) and benign (BPH-1) prostatic epithelial cell lines or silencing in the BPH-1 cell line, respectively, promoted or inhibited migration and spheroid invasion in collagen I gel and Matrigel. T-cadherin-dependent effects were associated with changes in cell phenotype: overexpression caused cell dissemination and loss of polarity evaluated by relative positioning of the Golgi/nuclei in cell groups, whereas silencing caused formation of compact polarized epithelial-like clusters. Epidermal growth factor receptor (EGFR) and IGF factor-1 receptor (IGF-1R) were identified as mediators of T-cadherin effects. These receptors per se had opposing influences on cell phenotype. EGFR activation with EGF or IGF-1R inhibition with NVP-AEW541 promoted dissemination, invasion, and polarity loss. Conversely, inhibition of EGFR with gefitinib or activation of IGF-1R with IGF-1 rescued epithelial morphology and decreased invasion. T-cadherin silencing enhanced both EGFR and IGF-1R phosphorylation, yet converted cells to the morphology typical for activated IGF-1R. T-cadherin effects were sensitive to modulation of EGFR or IGF-1R activity, suggesting direct involvement of both receptors. We conclude that T-cadherin regulates prostate cancer cell behavior by tuning the balance in EGFR/IGF-1R activity and enhancing the impact of IGF-1R.
Collapse
Affiliation(s)
- Kseniya Maslova
- *Department of Biomedicine, Laboratory for Signal Transduction, and Institute of Pathology, University Hospital Basel, University of Basel, Basel, Switzerland; and Hirslanden Klinik St. Anna, Luzern, Switzerland
| | - Emmanouil Kyriakakis
- *Department of Biomedicine, Laboratory for Signal Transduction, and Institute of Pathology, University Hospital Basel, University of Basel, Basel, Switzerland; and Hirslanden Klinik St. Anna, Luzern, Switzerland
| | - Dennis Pfaff
- *Department of Biomedicine, Laboratory for Signal Transduction, and Institute of Pathology, University Hospital Basel, University of Basel, Basel, Switzerland; and Hirslanden Klinik St. Anna, Luzern, Switzerland
| | - Audrey Frachet
- *Department of Biomedicine, Laboratory for Signal Transduction, and Institute of Pathology, University Hospital Basel, University of Basel, Basel, Switzerland; and Hirslanden Klinik St. Anna, Luzern, Switzerland
| | - Agne Frismantiene
- *Department of Biomedicine, Laboratory for Signal Transduction, and Institute of Pathology, University Hospital Basel, University of Basel, Basel, Switzerland; and Hirslanden Klinik St. Anna, Luzern, Switzerland
| | - Lukas Bubendorf
- *Department of Biomedicine, Laboratory for Signal Transduction, and Institute of Pathology, University Hospital Basel, University of Basel, Basel, Switzerland; and Hirslanden Klinik St. Anna, Luzern, Switzerland
| | - Christian Ruiz
- *Department of Biomedicine, Laboratory for Signal Transduction, and Institute of Pathology, University Hospital Basel, University of Basel, Basel, Switzerland; and Hirslanden Klinik St. Anna, Luzern, Switzerland
| | - Tatjana Vlajnic
- *Department of Biomedicine, Laboratory for Signal Transduction, and Institute of Pathology, University Hospital Basel, University of Basel, Basel, Switzerland; and Hirslanden Klinik St. Anna, Luzern, Switzerland
| | - Paul Erne
- *Department of Biomedicine, Laboratory for Signal Transduction, and Institute of Pathology, University Hospital Basel, University of Basel, Basel, Switzerland; and Hirslanden Klinik St. Anna, Luzern, Switzerland
| | - Thérèse J Resink
- *Department of Biomedicine, Laboratory for Signal Transduction, and Institute of Pathology, University Hospital Basel, University of Basel, Basel, Switzerland; and Hirslanden Klinik St. Anna, Luzern, Switzerland
| | - Maria Philippova
- *Department of Biomedicine, Laboratory for Signal Transduction, and Institute of Pathology, University Hospital Basel, University of Basel, Basel, Switzerland; and Hirslanden Klinik St. Anna, Luzern, Switzerland
| |
Collapse
|
32
|
Shenoy AK, Lu J. Cancer cells remodel themselves and vasculature to overcome the endothelial barrier. Cancer Lett 2014; 380:534-544. [PMID: 25449784 DOI: 10.1016/j.canlet.2014.10.031] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2014] [Revised: 10/27/2014] [Accepted: 10/28/2014] [Indexed: 12/19/2022]
Abstract
Metastasis refers to the spread of cancer cells from a primary tumor to distant organs mostly via the bloodstream. During the metastatic process, cancer cells invade blood vessels to enter circulation, and later exit the vasculature at a distant site. Endothelial cells that line blood vessels normally serve as a barrier to the movement of cells into or out of the blood. It is thus critical to understand how metastatic cancer cells overcome the endothelial barrier. Epithelial cancer cells acquire increased motility and invasiveness through epithelial-to-mesenchymal transition (EMT), which enables them to move toward vasculature. Cancer cells also express a variety of adhesion molecules that allow them to attach to vascular endothelium. Finally, cancer cells secrete or induce growth factors and cytokines to actively prompt vascular hyperpermeability that compromises endothelial barrier function and facilitates transmigration of cancer cells through the vascular wall. Elucidation of the mechanisms underlying metastatic dissemination may help develop new anti-metastasis therapeutics.
Collapse
Affiliation(s)
- Anitha K Shenoy
- Department of Biochemistry and Molecular Biology, University of Florida College of Medicine, Gainesville, FL 32610, United States.
| | - Jianrong Lu
- Department of Biochemistry and Molecular Biology, University of Florida College of Medicine, Gainesville, FL 32610, United States.
| |
Collapse
|
33
|
Nichols LA, Grunz-Borgmann EA, Wang X, Parrish AR. A role for the age-dependent loss of α(E)-catenin in regulation of N-cadherin expression and cell migration. Physiol Rep 2014; 2:2/6/e12039. [PMID: 24920123 PMCID: PMC4208646 DOI: 10.14814/phy2.12039] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
The aging kidney has a decreased ability to repair following acute kidney injury. Previous studies from our laboratory have demonstrated a loss in α‐catenin expression in the aging rat kidney. We hypothesize that loss of α‐catenin expression in tubular epithelial cells may induce changes that result in a decreased repair capacity. In these studies, we demonstrate that decreased α‐catenin protein expression is detectable as early as 20 months of age in male Fischer 344 rats. Protein loss is also observed in aged nonhuman primate kidneys, suggesting that this is not a species‐specific response. In an effort to elucidate alterations due to the loss of α‐catenin, we generated NRK‐52E cell lines with stable knockdown of α(E)‐catenin (C2 cells). Interestingly, C2 cells had decreased expression of N‐cadherin, decreased cell–cell adhesion, and increased monolayer permeability. C2 had deficits in wound repair, due to alterations in cell migration. Analysis of gene expression in the migrating control cells indicated that expression of N‐cadherin and N‐CAM was increased during repair. In migrating C2 cells, expression of N‐CAM was also increased, but the expression of N‐cadherin was not upregulated. Importantly, a blocking antibody against N‐cadherin inhibited repair in NRK‐52E cells, suggesting an important role in repair. Taken together, these data suggest that loss of α‐catenin, and the subsequent downregulation of N‐cadherin expression, is a mechanism underlying the decreased migration of tubular epithelial cells that contributes to the inability of the aging kidney to repair following injury. Aging is associated with loss of α‐catenin and N‐cadherin expression in the kidney. In these studies, we demonstrate that α‐catenin regulates, in part, N‐cadherin expression and migration in tubular epithelial cells.
Collapse
Affiliation(s)
- LaNita A Nichols
- Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia, Missouri
| | | | - Xinhui Wang
- Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia, Missouri
| | - Alan R Parrish
- Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia, Missouri
| |
Collapse
|