51
|
Hönig E, Ringer K, Dewes J, von Mach T, Kamm N, Kreitzer G, Jacob R. Galectin-3 modulates the polarized surface delivery of β1-integrin in epithelial cells. J Cell Sci 2018; 131:jcs.213199. [PMID: 29748377 DOI: 10.1242/jcs.213199] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Accepted: 04/30/2018] [Indexed: 12/25/2022] Open
Abstract
Epithelial cells require a precise intracellular transport and sorting machinery to establish and maintain their polarized architecture. This machinery includes β-galactoside-binding galectins for targeting of glycoprotein to the apical membrane. Galectin-3 sorts cargo destined for the apical plasma membrane into vesicular carriers. After delivery of cargo to the apical milieu, galectin-3 recycles back into sorting organelles. We analysed the role of galectin-3 in the polarized distribution of β1-integrin in MDCK cells. Integrins are located primarily at the basolateral domain of epithelial cells. We demonstrate that a minor pool of β1-integrin interacts with galectin-3 at the apical plasma membrane. Knockdown of galectin-3 decreases apical delivery of β1-integrin. This loss is restored by supplementation with recombinant galectin-3 and galectin-3 overexpression. Our data suggest that galectin-3 targets newly synthesized β1-integrin to the apical membrane and promotes apical delivery of β1-integrin internalized from the basolateral membrane. In parallel, knockout of galectin-3 results in a reduction in cell proliferation and an impairment in proper cyst development. Our results suggest that galectin-3 modulates the surface distribution of β1-integrin and affects the morphogenesis of polarized cells.
Collapse
Affiliation(s)
- Ellena Hönig
- Department of Cell Biology and Cell Pathology, Philipps-Universität Marburg, Marburg 35037, Germany
| | - Karina Ringer
- Department of Cell Biology and Cell Pathology, Philipps-Universität Marburg, Marburg 35037, Germany.,DFG Research Training Group, Membrane Plasticity in Tissue Development and Remodeling, GRK 2213, Philipps-Universität Marburg, Marburg 35043, Germany
| | - Jenny Dewes
- Department of Cell Biology and Cell Pathology, Philipps-Universität Marburg, Marburg 35037, Germany
| | - Tobias von Mach
- Department of Cell Biology and Cell Pathology, Philipps-Universität Marburg, Marburg 35037, Germany
| | - Natalia Kamm
- Department of Cell Biology and Cell Pathology, Philipps-Universität Marburg, Marburg 35037, Germany
| | - Geri Kreitzer
- Department of Molecular, Cellular and Biomedical Sciences, City University of New York School of Medicine, City College of New York, NY 10031, USA
| | - Ralf Jacob
- Department of Cell Biology and Cell Pathology, Philipps-Universität Marburg, Marburg 35037, Germany .,DFG Research Training Group, Membrane Plasticity in Tissue Development and Remodeling, GRK 2213, Philipps-Universität Marburg, Marburg 35043, Germany
| |
Collapse
|
52
|
Abstract
The concept that progression of cancer is regulated by interactions of cancer cells with their microenvironment was postulated by Stephen Paget over a century ago. Contemporary tumour microenvironment (TME) research focuses on the identification of tumour-interacting microenvironmental constituents, such as resident or infiltrating non-tumour cells, soluble factors and extracellular matrix components, and the large variety of mechanisms by which these constituents regulate and shape the malignant phenotype of tumour cells. In this Timeline article, we review the developmental phases of the TME paradigm since its initial description. While illuminating controversies, we discuss the importance of interactions between various microenvironmental components and tumour cells and provide an overview and assessment of therapeutic opportunities and modalities by which the TME can be targeted.
Collapse
Affiliation(s)
- Shelly Maman
- Department of Cell Research and Immunology, The George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Isaac P Witz
- Department of Cell Research and Immunology, The George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
53
|
Mussano F, Genova T, Petrillo S, Roato I, Ferracini R, Munaron L. Osteogenic Differentiation Modulates the Cytokine, Chemokine, and Growth Factor Profile of ASCs and SHED. Int J Mol Sci 2018; 19:ijms19051454. [PMID: 29757956 PMCID: PMC5983594 DOI: 10.3390/ijms19051454] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Revised: 05/11/2018] [Accepted: 05/11/2018] [Indexed: 02/06/2023] Open
Abstract
Great efforts have been made to improve bone regeneration techniques owing to a growing variety of sources of stem cells suitable for autologous transplants. Specifically, adipose-derived stem cells (ASCs) and stems cells from human exfoliated deciduous teeth (SHED) hold great potential for bone tissue engineering and cell therapy. After a preliminary characterization of the main biomolecules ASCs and SHED released in their conditioned media, cells were kept both in normal and osteo-inducing conditions. Conventional assays were performed to prove their osteogenic potential such as quantitative real-time polymerase chain reaction (qRT-PCR) (for RUNX-2, collagen type I, osteopontin and osteonectin), alkaline phosphatase activity, osteocalcin production, and von Kossa staining. Conditioned media were tested again after the osteogenic induction and compared to maintaining condition both at base line and after 14 days of culture. The osteogenic condition inhibited the release of all the biomolecules, with the exception, concerning SHED, of growth-regulated alpha protein precursor (GROα), and, to a lesser extent, interleukin (IL)-8. In conclusion, our data support that undifferentiated ASCs and SHED may be preferable to committed ones for general cell therapy approaches, due to their higher paracrine activity. Osteoinduction significantly affects the cytokine, chemokine, and growth factor profile in a differential way, as SHED kept a more pronounced pro-angiogenic signature than ASCs.
Collapse
Affiliation(s)
- Federico Mussano
- CIR Dental School, Department of Surgical Sciences UNITO, via Nizza 230, 10126 Turin, Italy.
| | - Tullio Genova
- CIR Dental School, Department of Surgical Sciences UNITO, via Nizza 230, 10126 Turin, Italy.
- Department of Life Sciences and Systems Biology, UNITO, via Accademia Albertina 13, 10123 Turin, Italy.
| | - Sara Petrillo
- Department of Molecular Biotechnology and Health Sciences, UNITO, Via Nizza 52, 10126 Turin, Italy.
| | - Ilaria Roato
- Center for Research and Medical Studies, A.O.U. Città della Salute e della Scienza, 10126 Turin, Italy.
| | - Riccardo Ferracini
- Department of Surgical Sciences (DISC), Orthopaedic Clinic-IRCCS A.O.U. San Martino, 16132 Genoa, Italy.
| | - Luca Munaron
- Department of Life Sciences and Systems Biology, UNITO, via Accademia Albertina 13, 10123 Turin, Italy.
| |
Collapse
|
54
|
Mehta S, Lo Cascio C. Developmentally regulated signaling pathways in glioma invasion. Cell Mol Life Sci 2018; 75:385-402. [PMID: 28821904 PMCID: PMC5765207 DOI: 10.1007/s00018-017-2608-8] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Revised: 07/18/2017] [Accepted: 08/03/2017] [Indexed: 01/06/2023]
Abstract
Malignant gliomas are the most common, infiltrative, and lethal primary brain tumors affecting the adult population. The grim prognosis for this disease is due to a combination of the presence of highly invasive tumor cells that escape surgical resection and the presence of a population of therapy-resistant cancer stem cells found within these tumors. Several studies suggest that glioma cells have cleverly hijacked the normal developmental program of neural progenitor cells, including their transcriptional programs, to enhance gliomagenesis. In this review, we summarize the role of developmentally regulated signaling pathways that have been found to facilitate glioma growth and invasion. Furthermore, we discuss how the microenvironment and treatment-induced perturbations of these highly interconnected signaling networks can trigger a shift in cellular phenotype and tumor subtype.
Collapse
Affiliation(s)
- Shwetal Mehta
- Division of Neurobiology, Barrow Brain Tumor Research Center, Barrow Neurological Institute, Phoenix, AZ, 85013, USA.
| | - Costanza Lo Cascio
- Division of Neurobiology, Barrow Brain Tumor Research Center, Barrow Neurological Institute, Phoenix, AZ, 85013, USA
| |
Collapse
|
55
|
Yamaji D, Soliman MM, Kamikawa A, Ito T, Ahmed MM, Okamatsu-Ogura Y, Saito M, Kimura K. Species-specific control of hepatocyte growth factor expression and production in adipocytes in a differentiation-dependent manner. Domest Anim Endocrinol 2018; 62:39-48. [PMID: 29040876 DOI: 10.1016/j.domaniend.2017.09.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Revised: 08/22/2017] [Accepted: 09/06/2017] [Indexed: 01/09/2023]
Abstract
Hepatocyte growth factor (HGF) is a mesenchymal cell-derived factor that regulates cell growth, cell motility, and morphogenesis. Since there are conflicting reports on HGF-producing cells, we herein examined HGF activity in conditioned medium (CM) of bovine and mouse preadipocytes before and after adipogenic differentiation. CM of bovine adipocytes and mouse preadipocytes induced the morphogenesis of mammary epithelial cells that was inhibited by an NK4 HGF antagonist, whereas CM of bovine preadipocytes and mouse adipocytes did not. HGF mRNA expression was increased by a treatment with dexamethasone and isobutylmethylxanthine in bovine as well as human cells, whereas it was decreased in rodent cells. It was unfortunate that HGF gene promoter activity failed to reflect HGF mRNA expression in these cells. After actinomycin D treatment, expression of HGF mRNA remained stable in pre- and differentiated bovine adipocytes and mouse preadipocytes, whereas rapidly decreased in mouse-differentiated adipocytes. These results indicate that expression and production of HGF are regulated in a species-specific adipogenic differentiation-dependent manner and suggest that the decrease in HGF mRNA in mouse differentiated adipocytes is, at least in part, mediated by differentiation-dependent loss of its stability.
Collapse
Affiliation(s)
- D Yamaji
- Laboratory of Biochemistry, Department of Biomedical Sciences, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo 060-0818, Japan
| | - M M Soliman
- Laboratory of Biochemistry, Department of Biomedical Sciences, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo 060-0818, Japan
| | - A Kamikawa
- Laboratory of Biochemistry, Department of Biomedical Sciences, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo 060-0818, Japan
| | - T Ito
- Laboratory of Biochemistry, Department of Biomedical Sciences, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo 060-0818, Japan
| | - M M Ahmed
- Laboratory of Biochemistry, Department of Biomedical Sciences, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo 060-0818, Japan
| | - Y Okamatsu-Ogura
- Laboratory of Biochemistry, Department of Biomedical Sciences, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo 060-0818, Japan
| | - M Saito
- Laboratory of Biochemistry, Department of Biomedical Sciences, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo 060-0818, Japan
| | - K Kimura
- Laboratory of Biochemistry, Department of Biomedical Sciences, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo 060-0818, Japan.
| |
Collapse
|
56
|
Nishimura R, Kato K, Fujiwara S, Ohashi K, Mizuno K. Solo and Keratin Filaments Regulate Epithelial Tubule Morphology. Cell Struct Funct 2018; 43:95-105. [DOI: 10.1247/csf.18010] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Affiliation(s)
- Ryosuke Nishimura
- Department of Biomolecular Sciences, Graduate School of Life Sciences, Tohoku University
- Department of Cell Biology, Graduate School of Medical Sciences, Tokushima University
| | - Kagayaki Kato
- Bioimage Informatics Group, Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Sciences (NINS)
- Department of Imaging Science, Center for Novel Science Initiatives (CNSI), National Institutes of Natural Sciences (NINS)
- Division of Evolutionary Biology Biodiversity, National Institute for Basic Biology (NIBB)
| | - Sachiko Fujiwara
- Division of Bioengineering, Graduate School of Engineering Science, Osaka University
| | - Kazumasa Ohashi
- Department of Biomolecular Sciences, Graduate School of Life Sciences, Tohoku University
| | - Kensaku Mizuno
- Department of Biomolecular Sciences, Graduate School of Life Sciences, Tohoku University
| |
Collapse
|
57
|
Miyagi H, Thomasy SM, Russell P, Murphy CJ. The role of hepatocyte growth factor in corneal wound healing. Exp Eye Res 2018; 166:49-55. [PMID: 29024692 PMCID: PMC5831200 DOI: 10.1016/j.exer.2017.10.006] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Revised: 09/19/2017] [Accepted: 10/08/2017] [Indexed: 11/28/2022]
Abstract
Hepatocyte growth factor (HGF) is a glycoprotein produced by mesenchymal cells and operates as a key molecule for tissue generation and renewal. During corneal injury, HGF is primarily secreted by stromal fibroblasts and promotes epithelial wound healing in a paracrine manner. While this mesenchymal-epithelial interaction is well characterized in various organs and the cornea, the role of HGF in corneal stromal and endothelial wound healing is understudied. In addition, HGF has been shown to play an anti-fibrotic role by inhibiting myofibroblast generation and subsequent production of a disorganized extracellular matrix and tissue fibrosis. Therefore, HGF represents a potential therapeutic tool in numerous organs in which myofibroblasts are responsible for tissue scarring. Corneal fibrosis can be a devastating sequela of injury and can result in corneal opacification and retrocorneal membrane formation leading to severe vision loss. In this article, we concisely review the available literature regarding the role of HGF in corneal wound healing. We highlight the influence of HGF on cellular behaviors in each corneal layer. Additionally, we suggest the possibility that HGF may represent a therapeutic tool for interrupting dysregulated corneal repair processes to improve patient outcomes.
Collapse
Affiliation(s)
- Hidetaka Miyagi
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California, 1 Shields Ave., Davis, CA, 95616, USA; Department of Ophthalmology and Visual Sciences, Graduate School of Biomedical Sciences, Hiroshima University, Minami-ku, Kasumi 1-2-3, Hiroshima, 7348551, Japan.
| | - Sara M Thomasy
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California, 1 Shields Ave., Davis, CA, 95616, USA; Department of Ophthalmology & Vision Science, School of Medicine, UC Davis Medical Center, 2315 Stockton Blvd, Sacramento, CA, 95817, USA.
| | - Paul Russell
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California, 1 Shields Ave., Davis, CA, 95616, USA.
| | - Christopher J Murphy
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California, 1 Shields Ave., Davis, CA, 95616, USA; Department of Ophthalmology & Vision Science, School of Medicine, UC Davis Medical Center, 2315 Stockton Blvd, Sacramento, CA, 95817, USA.
| |
Collapse
|
58
|
Activated HGF-c-Met Axis in Head and Neck Cancer. Cancers (Basel) 2017; 9:cancers9120169. [PMID: 29231907 PMCID: PMC5742817 DOI: 10.3390/cancers9120169] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Revised: 12/06/2017] [Accepted: 12/07/2017] [Indexed: 12/14/2022] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC) is a highly morbid disease. Recent developments including Food and Drug Administration (FDA) approved molecular targeted agent’s pembrolizumab and cetuximab show promise but did not improve the five-year survival which is currently less than 40%. The hepatocyte growth factor receptor; also known as mesenchymal–epithelial transition factor (c-Met) and its ligand hepatocyte growth factor (HGF) are overexpressed in head and neck squamous cell carcinoma (HNSCC); and regulates tumor progression and response to therapy. The c-Met pathway has been shown to regulate many cellular processes such as cell proliferation, invasion, and angiogenesis. The c-Met pathway is involved in cross-talk, activation, and perpetuation of other signaling pathways, curbing the cogency of a blockade molecule on a single pathway. The receptor and its ligand act on several downstream effectors including phospholipase C gamma (PLCγ), cellular Src kinase (c-Src), phosphotidylinsitol-3-OH kinase (PI3K) alpha serine/threonine-protein kinase (Akt), mitogen activate protein kinase (MAPK), and wingless-related integration site (Wnt) pathways. They are also known to cross-talk with other receptors; namely epidermal growth factor receptor (EGFR) and vascular endothelial growth factor receptor (VEGFR) and specifically contribute to treatment resistance. Clinical trials targeting the c-Met axis in HNSCC have been undertaken because of significant preclinical work demonstrating a relationship between HGF/c-Met signaling and cancer cell survival. Here we focus on HGF/c-Met impact on cellular signaling in HNSCC to potentiate tumor growth and disrupt therapeutic efficacy. Herein we summarize the current understanding of HGF/c-Met signaling and its effects on HNSCC. The intertwining of c-Met signaling with other signaling pathways provides opportunities for more robust and specific therapies, leading to better clinical outcomes.
Collapse
|
59
|
Hirashima T, Hoshuyama M, Adachi T. In vitro tubulogenesis of Madin–Darby canine kidney (MDCK) spheroids occurs depending on constituent cell number and scaffold gel concentration. J Theor Biol 2017; 435:110-115. [DOI: 10.1016/j.jtbi.2017.09.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2017] [Revised: 08/23/2017] [Accepted: 09/11/2017] [Indexed: 02/06/2023]
|
60
|
Xi W, Sonam S, Beng Saw T, Ladoux B, Teck Lim C. Emergent patterns of collective cell migration under tubular confinement. Nat Commun 2017; 8:1517. [PMID: 29142242 PMCID: PMC5688140 DOI: 10.1038/s41467-017-01390-x] [Citation(s) in RCA: 93] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Accepted: 09/14/2017] [Indexed: 02/07/2023] Open
Abstract
Collective epithelial behaviors are essential for the development of lumens in organs. However, conventional assays of planar systems fail to replicate cell cohorts of tubular structures that advance in concerted ways on out-of-plane curved and confined surfaces, such as ductal elongation in vivo. Here, we mimic such coordinated tissue migration by forming lumens of epithelial cell sheets inside microtubes of 1-10 cell lengths in diameter. We show that these cell tubes reproduce the physiological apical-basal polarity, and have actin alignment, cell orientation, tissue organization, and migration modes that depend on the extent of tubular confinement and/or curvature. In contrast to flat constraint, the cell sheets in a highly constricted smaller microtube demonstrate slow motion with periodic relaxation, but fast overall movement in large microtubes. Altogether, our findings provide insights into the emerging migratory modes for epithelial migration and growth under tubular confinement, which are reminiscent of the in vivo scenario.
Collapse
Affiliation(s)
- Wang Xi
- Mechanobiology Institute, National University of Singapore, 5A Engineering Drive 1, Singapore, 117411, Singapore
- Centre for Advanced 2D Materials and Graphene Research Centre, National University of Singapore, 6 Science Drive 2, Singapore, 117546, Singapore
| | - Surabhi Sonam
- Mechanobiology Institute, National University of Singapore, 5A Engineering Drive 1, Singapore, 117411, Singapore
- Department of Biomedical Engineering and Department of Mechanical Engineering, National University of Singapore, Singapore, 117575, Singapore
- Institut Jacques Monod, Université Paris Diderot & CNRS UMR 7592, 75205, Paris cedex 13, France
| | - Thuan Beng Saw
- Mechanobiology Institute, National University of Singapore, 5A Engineering Drive 1, Singapore, 117411, Singapore
- NUS Graduate School of Integrative Sciences and Engineering, National University of Singapore, Singapore, 117456, Singapore
| | - Benoit Ladoux
- Mechanobiology Institute, National University of Singapore, 5A Engineering Drive 1, Singapore, 117411, Singapore.
- Institut Jacques Monod, Université Paris Diderot & CNRS UMR 7592, 75205, Paris cedex 13, France.
| | - Chwee Teck Lim
- Mechanobiology Institute, National University of Singapore, 5A Engineering Drive 1, Singapore, 117411, Singapore.
- Centre for Advanced 2D Materials and Graphene Research Centre, National University of Singapore, 6 Science Drive 2, Singapore, 117546, Singapore.
- Department of Biomedical Engineering and Department of Mechanical Engineering, National University of Singapore, Singapore, 117575, Singapore.
- NUS Graduate School of Integrative Sciences and Engineering, National University of Singapore, Singapore, 117456, Singapore.
- Biomedical Institute for Global Health Research and Technology, National University of Singapore, #14-01, MD6, 14 Medical Drive, Singapore, 117599, Singapore.
| |
Collapse
|
61
|
Ma J, Sun X, Guo T, Su H, Chen Q, Gong Z, Qi J, Zhao X. Interleukin-1 receptor antagonist inhibits angiogenesis via blockage IL-1α/PI3K/NF-κβ pathway in human colon cancer cell. Cancer Manag Res 2017; 9:481-493. [PMID: 29062241 PMCID: PMC5640395 DOI: 10.2147/cmar.s147699] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
PURPOSE This article investigates the relationship between cancer cells and stromal cells in carcinoma cell living microenvironment and elucidates the mechanism that interleukin-1 receptor antagonist (IL-1RA) blocks metastatic potential in colon cancer. METHODS Western blot and RT-PCR assay were used to determine the expression of hepatocyte growth factor (HGF) and IL-1α in colon carcinoma cells and stromal cells. Effect of IL-1RA and HGF on metastatic potential of colon cancer cells were examined by proliferation, invasion, and angiogenesis assays. The interactional role of IL-1RA and HGF were detected by ELISA assay, invasion, and angiogenesis assay making up a co-culture system consisting of stromal and colon cancer cells in cells living microenvironment. RESULTS IL-1α was expressed in human umbilical vein endothelial cells (HUVECs) and HT-29 and WiDr (colon cancer cell lines with higher liver metastatic potential). HGF was expressed only in fibroblast. HGF secretion from fibroblasts was significantly inhibited by IL-1RA (P<0.01). Furthermore, IL-1RA could significantly inhibit migration, proliferation, and angiogenesis of HUVECs (P<0.01). In the double co-culture system, there is a high liver metastatic potential of colon cancer cell line (HT-29) because it can secrete autocrine IL-1α, significantly enhanced angiogenesis compared with low liver metastatic cell line (CaCo-2) (P<0.01), which does not secrete IL-1α. On the contrary, blockage of autocrine IL-1α by IL-1RA might significantly decrease metastatic potential of colon carcinoma cells through downregulation of IL-1α/PI3K/NF-κB pathway. CONCLUSION IL-1 receptor antagonist (IL-1RA) is an important inhibitor in metastatic process of colon carcinoma cell. Based on the above results, we suggest that IL-1RA may be a promising new therapeutic approach in inhibiting colon cancer with IL-1-producing patients.
Collapse
Affiliation(s)
- Jiachi Ma
- Department of General Surgery, Gansu Provincial People’s Hospital, LanZhou
| | - Xiaowen Sun
- Department of Dermatology, The First Hospital of Tianshui, Tianshui
| | - Tiankang Guo
- Department of General Surgery, Gansu Provincial People’s Hospital, LanZhou
| | - He Su
- Department of General Surgery, Gansu Provincial People’s Hospital, LanZhou
| | - Quan Chen
- Department of General Surgery, Gansu Provincial People’s Hospital, LanZhou
| | - Zhenqiang Gong
- Ningxia Medical University Graduate School of Medical Sciences, Surgical Oncology, Yinchuan, People’s Republic of China
| | - Jianbo Qi
- Ningxia Medical University Graduate School of Medical Sciences, Surgical Oncology, Yinchuan, People’s Republic of China
| | - Xiaodan Zhao
- Ningxia Medical University Graduate School of Medical Sciences, Surgical Oncology, Yinchuan, People’s Republic of China
| |
Collapse
|
62
|
Schmutz CF, Hatton TD, Blomfield MP, Chandler BD, Benedick TC, Hansen MDH. A cellular automaton based on plasma membrane turnover accurately recapitulates cell mechanics during epithelial scattering. J Theor Biol 2017. [PMID: 28648562 DOI: 10.1016/j.jtbi.2017.06.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Epithelial cells can be triggered to actively detach from epithelial tissues and become solitary, migratory and invasive. This process occurs repeatedly in development, where it is termed epithelial-mesenchymal transition (EMT), and can be recapitulated as epithelial scattering in cell culture models. Detachment of cell-cell junctions involves changes in contractile forces, actin cytoskeletal organization, changes in cell-substrate adhesion properties, surface presentation of cell-cell adhesion molecules, and gene expression. That these cellular processes affect each other and share molecular components creates difficulties in generating hypotheses and designing experiments to understand the mechanics of epithelial scattering. Computational modeling is proving a powerful too in such instances. Here we develop a cellular automaton to reveal insights into how cells rupture epithelial cell-cell junctions during scattering. The model is optimized for realistic and stable recapitulation of behavior of single cells, then for realistic simulation of multiple cells forming epithelial colonies. With a workable model of epithelial cell behavior, we then alter model parameters and assess whether we can realistically mimic epithelial scattering. Adjusting model parameters to recapitulate epithelial scattering reveals that induction of cell migration is the major driver of epithelial scattering.
Collapse
Affiliation(s)
- Christian F Schmutz
- Physiology and Developmental Biology, Brigham Young University, 4005 LSB, Provo, UT 84602, USA
| | - Trey D Hatton
- Physiology and Developmental Biology, Brigham Young University, 4005 LSB, Provo, UT 84602, USA
| | - Michael P Blomfield
- Physiology and Developmental Biology, Brigham Young University, 4005 LSB, Provo, UT 84602, USA
| | - Benjamin D Chandler
- Physiology and Developmental Biology, Brigham Young University, 4005 LSB, Provo, UT 84602, USA
| | - Trent C Benedick
- Physiology and Developmental Biology, Brigham Young University, 4005 LSB, Provo, UT 84602, USA
| | - Marc D H Hansen
- Physiology and Developmental Biology, Brigham Young University, 4005 LSB, Provo, UT 84602, USA.
| |
Collapse
|
63
|
Tyrosine dephosphorylated cortactin downregulates contractility at the epithelial zonula adherens through SRGAP1. Nat Commun 2017; 8:790. [PMID: 28983097 PMCID: PMC5629210 DOI: 10.1038/s41467-017-00797-w] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2016] [Accepted: 07/20/2017] [Indexed: 11/25/2022] Open
Abstract
Contractile adherens junctions support cell−cell adhesion, epithelial integrity, and morphogenesis. Much effort has been devoted to understanding how contractility is established; however, less is known about whether contractility can be actively downregulated at junctions nor what function this might serve. We now identify such an inhibitory pathway that is mediated by the cytoskeletal scaffold, cortactin. Mutations of cortactin that prevent its tyrosine phosphorylation downregulate RhoA signaling and compromise the ability of epithelial cells to generate a contractile zonula adherens. This is mediated by the RhoA antagonist, SRGAP1. We further demonstrate that this mechanism is co-opted by hepatocyte growth factor to promote junctional relaxation and motility in epithelial collectives. Together, our findings identify a novel function of cortactin as a regulator of RhoA signaling that can be utilized by morphogenetic regulators for the active downregulation of junctional contractility. Epithelial cell-cell adhesions are contractile junctions, but whether contractility can be down-regulated is not known. Here the authors report how tyrosine dephosphorylation of the cytoskeletal scaffold, cortactin, recruits the RhoA antagonist SRGAP1 to relax adherens junctions in response to HGF.
Collapse
|
64
|
Datta A, Sandilands E, Mostov KE, Bryant DM. Fibroblast-derived HGF drives acinar lung cancer cell polarization through integrin-dependent RhoA-ROCK1 inhibition. Cell Signal 2017; 40:91-98. [PMID: 28888686 DOI: 10.1016/j.cellsig.2017.09.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Revised: 08/24/2017] [Accepted: 09/04/2017] [Indexed: 12/14/2022]
Abstract
The formation of lumens in epithelial tissues requires apical-basal polarization of cells, and the co-ordination of this individual polarity collectively around a contiguous lumen. Signals from the Extracellular Matrix (ECM) instruct epithelia as to the orientation of where basal, and thus consequently apical, surfaces should be formed. We report that this pathway is normally absent in Calu-3 human lung adenocarcinoma cells in 3-Dimensional culture, but that paracrine signals from MRC5 lung fibroblasts can induce correct orientation of polarity and acinar morphogenesis. We identify HGF, acting through the c-Met receptor, as the key polarity-inducing morphogen, which acts to activate β1-integrin-dependent adhesion. HGF and ECM-derived integrin signals co-operate via a c-Src-dependent inhibition of the RhoA-ROCK1 signalling pathway via p190A RhoGAP. This occurred via controlling localization of these signalling pathways to the ECM-abutting surface of cells in 3-Dimensional culture. Thus, stromal derived signals can influence morphogenesis in epithelial cells by controlling activation and localization of cell polarity pathways.
Collapse
Affiliation(s)
- Anirban Datta
- Dept. of Anatomy, University of California San Francisco, CA 94158-2140, USA; Dept. of Biochemistry and Biophysics, University of California San Francisco, CA 94158-2140, USA
| | - Emma Sandilands
- Institute of Cancer Sciences, University of Glasgow, Glasgow G61 1BD, United Kingdom
| | - Keith E Mostov
- Dept. of Anatomy, University of California San Francisco, CA 94158-2140, USA; Dept. of Biochemistry and Biophysics, University of California San Francisco, CA 94158-2140, USA
| | - David M Bryant
- Institute of Cancer Sciences, University of Glasgow, Glasgow G61 1BD, United Kingdom; The CRUK Beatson Institute, Glasgow G61 1BD, United Kingdom.
| |
Collapse
|
65
|
Weber HM, Tsurkan MV, Magno V, Freudenberg U, Werner C. Heparin-based hydrogels induce human renal tubulogenesis in vitro. Acta Biomater 2017; 57:59-69. [PMID: 28526628 DOI: 10.1016/j.actbio.2017.05.035] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Revised: 05/03/2017] [Accepted: 05/15/2017] [Indexed: 12/01/2022]
Abstract
Dialysis or kidney transplantation is the only therapeutic option for end stage renal disease. Accordingly, there is a large unmet clinical need for new causative therapeutic treatments. Obtaining robust models that mimic the complex nature of the human kidney is a critical step in the development of new therapeutic strategies. Here we establish a synthetic in vitro human renal tubulogenesis model based on a tunable glycosaminoglycan-hydrogel platform. In this system, renal tubulogenesis can be modulated by the adjustment of hydrogel mechanics and degradability, growth factor signaling, and the presence of insoluble adhesion cues, potentially providing new insights for regenerative therapy. Different hydrogel properties were systematically investigated for their ability to regulate renal tubulogenesis. Hydrogels based on heparin and matrix metalloproteinase cleavable peptide linker units were found to induce the morphogenesis of single human proximal tubule epithelial cells into physiologically sized tubule structures. The generated tubules display polarization markers, extracellular matrix components, and organic anion transport functions of the in vivo renal proximal tubule and respond to nephrotoxins comparable to the human clinical response. The established hydrogel-based human renal tubulogenesis model is thus considered highly valuable for renal regenerative medicine and personalized nephrotoxicity studies. STATEMENT OF SIGNIFICANCE The only cure for end stage kidney disease is kidney transplantation. Hence, there is a huge need for reliable human kidney models to study renal regeneration and establish alternative treatments. Here we show the development and application of an in vitro human renal tubulogenesis model using heparin-based hydrogels. To the best of our knowledge, this is the first system where human renal tubulogenesis can be monitored from single cells to physiologically sized tubule structures in a tunable hydrogel system. To validate the efficacy of our model as a drug toxicity platform, a chemotherapy drug was incubated with the model, resulting in a drug response similar to human clinical pathology. The established model could have wide applications in the field of nephrotoxicity and renal regenerative medicine and offer a reliable alternative to animal models.
Collapse
Affiliation(s)
- Heather M Weber
- Leibniz Institute of Polymer Research Dresden, Max Bergmann Center of Biomaterials Dresden, Hohe Straße 6, 01069 Dresden, Germany.
| | - Mikhail V Tsurkan
- Leibniz Institute of Polymer Research Dresden, Max Bergmann Center of Biomaterials Dresden, Hohe Straße 6, 01069 Dresden, Germany.
| | - Valentina Magno
- Leibniz Institute of Polymer Research Dresden, Max Bergmann Center of Biomaterials Dresden, Hohe Straße 6, 01069 Dresden, Germany.
| | - Uwe Freudenberg
- Leibniz Institute of Polymer Research Dresden, Max Bergmann Center of Biomaterials Dresden, Hohe Straße 6, 01069 Dresden, Germany.
| | - Carsten Werner
- Leibniz Institute of Polymer Research Dresden, Max Bergmann Center of Biomaterials Dresden, Hohe Straße 6, 01069 Dresden, Germany; Center for Regenerative Therapies Dresden, Technische Universität Dresden, Fetscherstrasse 105, 01307 Dresden, Germany.
| |
Collapse
|
66
|
Ernandez T, Komarynets O, Chassot A, Sougoumarin S, Soulié P, Wang Y, Montesano R, Feraille E. Primary cilia control the maturation of tubular lumen in renal collecting duct epithelium. Am J Physiol Cell Physiol 2017; 313:C94-C107. [DOI: 10.1152/ajpcell.00290.2016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Revised: 04/28/2017] [Accepted: 04/28/2017] [Indexed: 11/22/2022]
Abstract
The key role of the primary cilium in developmental processes is illustrated by ciliopathies resulting from genetic defects of its components. Ciliopathies include a large variety of dysmorphic syndromes that share in common the presence of multiple kidney cysts. These observations suggest that primary cilia may control morphogenetic processes in the developing kidney. In this study, we assessed the role of primary cilium in branching tubulogenesis and/or lumen development using kidney collecting duct-derived mCCDN21 cells that display spontaneous tubulogenic properties when grown in collagen-Matrigel matrix. Tubulogenesis and branching were not altered when cilium body growth was inhibited by Kif3A or Ift88 silencing. In agreement with the absence of a morphogenetic effect, proliferation and wound-healing assay revealed that neither cell proliferation nor migration were altered by cilium body disruption. The absence of cilium following Kif3A or Ift88 silencing in mCCDN21 cells did not alter the initial stages of tubular lumen generation while lumen maturation and enlargement were delayed. This delay in tubular lumen maturation was not observed after Pkd1 knockdown in mCCDN21 cells. The delayed lumen maturation was explained by neither defective secretion or increased reabsorption of luminal fluid. Our results indicate that primary cilia do not control early morphogenetic processes in renal epithelium. Rather, primary cilia modulate tubular lumen maturation and enlargement resulting from luminal fluid accumulation in tubular structures derived from collecting duct cells.
Collapse
Affiliation(s)
- Thomas Ernandez
- Service of Nephrology, University Hospital of Geneva, Geneva, Switzerland; and
| | - Olga Komarynets
- Department of Cell Physiology and Metabolism, University Medical Center, Geneva, Switzerland
| | - Alexandra Chassot
- Department of Cell Physiology and Metabolism, University Medical Center, Geneva, Switzerland
| | - Soushma Sougoumarin
- Department of Cell Physiology and Metabolism, University Medical Center, Geneva, Switzerland
| | - Priscilla Soulié
- Department of Cell Physiology and Metabolism, University Medical Center, Geneva, Switzerland
| | - Yubao Wang
- Department of Cell Physiology and Metabolism, University Medical Center, Geneva, Switzerland
| | - Roberto Montesano
- Department of Cell Physiology and Metabolism, University Medical Center, Geneva, Switzerland
| | - Eric Feraille
- Service of Nephrology, University Hospital of Geneva, Geneva, Switzerland; and
- Department of Cell Physiology and Metabolism, University Medical Center, Geneva, Switzerland
| |
Collapse
|
67
|
Huling J, Yoo JJ. Comparing adult renal stem cell identification, characterization and applications. J Biomed Sci 2017; 24:32. [PMID: 28511675 PMCID: PMC5434527 DOI: 10.1186/s12929-017-0339-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Accepted: 05/03/2017] [Indexed: 12/27/2022] Open
Abstract
Despite growing interest and effort, a consensus has yet to be reached in regards to the identification of adult renal stem cells. Organ complexity and low turnover of renal cells has made stem cell identification difficult and lead to the investigation of multiple possible populations. In this review, we summarize the work that has been done toward finding and characterizing an adult renal stem cell population. In addition to giving a general overview of what has been done, we aim to highlight the variation in methods and outcomes. The methods used to locate potential stem cell populations can vary widely, but even within the relatively standard practice of BrdU labeling of slowly dividing cells, there are significant differences in protocols and results. Additional diversity exists in cell marker profiles and apparent differentiation potential seen in potential stem cell sources. Cataloging the variety of methods and outcomes seen so far may help to streamline future investigation and stear the field toward consensus. But even without firmly defined populations, the application of renal stem cells holds tantalizing potential. Populations of highly proliferative, multipotent cells of renal origin show the ability to engraft in injured kidneys, mitigate functional loss and occasionally show the ability to generate nephrons de novo. The progress toward regenerative medicine applications is also summarized.
Collapse
Affiliation(s)
- Jennifer Huling
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, 27157, USA.
| | - James J Yoo
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, 27157, USA
| |
Collapse
|
68
|
Omoto M, Suri K, Amouzegar A, Li M, Katikireddy KR, Mittal SK, Chauhan SK. Hepatocyte Growth Factor Suppresses Inflammation and Promotes Epithelium Repair in Corneal Injury. Mol Ther 2017; 25:1881-1888. [PMID: 28502469 DOI: 10.1016/j.ymthe.2017.04.020] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Revised: 04/11/2017] [Accepted: 04/17/2017] [Indexed: 10/19/2022] Open
Abstract
Corneal injuries are among the major causes of ocular morbidity and vision impairment. Optimal epithelial wound healing is critical for the integrity and transparency of the cornea after injury. Hepatocyte growth factor (HGF) is a mitogen and motility factor that primarily regulates epithelial cell function. Herein, we investigate the effect of HGF on proliferation of corneal epithelial cells (CECs) in inflamed conditions both in vitro and in vivo. We demonstrate that HGF not only promotes CEC proliferation in homeostatic conditions but also reverses the anti-proliferative effect of the inflammatory environment on these cells. Furthermore, using a mouse model of ocular injury, we show that HGF treatment suppresses ocular inflammation and actively augments CEC proliferation, leading to improved and accelerated corneal epithelial repair. These findings have potential translational implications and could provide a framework for the development of novel HGF-based therapies for corneal epithelial defects.
Collapse
Affiliation(s)
- Masahiro Omoto
- Schepens Eye Research Institute, Massachusetts Eye and Ear Infirmary, Boston, MA 02114, USA; Department of Ophthalmology, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Kunal Suri
- Schepens Eye Research Institute, Massachusetts Eye and Ear Infirmary, Boston, MA 02114, USA; Department of Ophthalmology, Harvard Medical School, Boston, MA 02115, USA
| | - Afsaneh Amouzegar
- Schepens Eye Research Institute, Massachusetts Eye and Ear Infirmary, Boston, MA 02114, USA; Department of Ophthalmology, Harvard Medical School, Boston, MA 02115, USA
| | - Mingshun Li
- Schepens Eye Research Institute, Massachusetts Eye and Ear Infirmary, Boston, MA 02114, USA; Department of Ophthalmology, Harvard Medical School, Boston, MA 02115, USA
| | - Kishore R Katikireddy
- Schepens Eye Research Institute, Massachusetts Eye and Ear Infirmary, Boston, MA 02114, USA; Department of Ophthalmology, Harvard Medical School, Boston, MA 02115, USA
| | - Sharad K Mittal
- Schepens Eye Research Institute, Massachusetts Eye and Ear Infirmary, Boston, MA 02114, USA; Department of Ophthalmology, Harvard Medical School, Boston, MA 02115, USA
| | - Sunil K Chauhan
- Schepens Eye Research Institute, Massachusetts Eye and Ear Infirmary, Boston, MA 02114, USA; Department of Ophthalmology, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
69
|
Matsumoto K, Umitsu M, De Silva DM, Roy A, Bottaro DP. Hepatocyte growth factor/MET in cancer progression and biomarker discovery. Cancer Sci 2017; 108:296-307. [PMID: 28064454 PMCID: PMC5378267 DOI: 10.1111/cas.13156] [Citation(s) in RCA: 174] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2016] [Revised: 12/26/2016] [Accepted: 01/03/2017] [Indexed: 02/06/2023] Open
Abstract
Signaling driven by hepatocyte growth factor (HGF) and MET receptor facilitates conspicuous biological responses such as epithelial cell migration, 3‐D morphogenesis, and survival. The dynamic migration and promotion of cell survival induced by MET activation are bases for invasion–metastasis and resistance, respectively, against targeted drugs in cancers. Recent studies indicated that MET in tumor‐derived exosomes facilitates metastatic niche formation and metastasis in malignant melanoma. In lung cancer, gene amplification‐induced MET activation and ligand‐dependent MET activation in an autocrine/paracrine manner are causes for resistance to epidermal growth factor receptor tyrosine kinase inhibitors and anaplastic lymphoma kinase inhibitors. Hepatocyte growth factor secreted in the tumor microenvironment contributes to the innate and acquired resistance to RAF inhibitors. Changes in serum/plasma HGF, soluble MET (sMET), and phospho‐MET have been confirmed to be associated with disease progression, metastasis, therapy response, and survival. Higher serum/plasma HGF levels are associated with therapy resistance and/or metastasis, while lower HGF levels are associated with progression‐free survival and overall survival after treatment with targeted drugs in lung cancer, gastric cancer, colon cancer, and malignant melanoma. Urinary sMET levels in patients with bladder cancer are higher than those in patients without bladder cancer and associated with disease progression. Some of the multi‐kinase inhibitors that target MET have received regulatory approval, whereas none of the selective HGF‐MET inhibitors have shown efficacy in phase III clinical trials. Validation of the HGF‐MET pathway as a critical driver in cancer development/progression and utilization of appropriate biomarkers are key to development and approval of HGF‐MET inhibitors for clinical use.
Collapse
Affiliation(s)
- Kunio Matsumoto
- Division of Tumor Dynamics, Cancer Research Institute, Kanazawa University, Kanazawa, Japan
| | - Masataka Umitsu
- Laboratory of Protein Synthesis and Expression, Institute for Protein Research, Osaka University, Osaka, Japan
| | - Dinuka M De Silva
- Urologic Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Arpita Roy
- Urologic Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Donald P Bottaro
- Urologic Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
70
|
Farahat M, Sathi GA, Hara ES, Taketa H, Kuboki T, Matsumoto T. MSCs feeder layers induce SMG self-organization and branching morphogenesis. PLoS One 2017; 12:e0176453. [PMID: 28448600 PMCID: PMC5407632 DOI: 10.1371/journal.pone.0176453] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Accepted: 04/11/2017] [Indexed: 01/14/2023] Open
Abstract
Dysfunction of salivary glands leads to several oral health problems, including dental caries, mastication and swallowing dysfunctions and multiple oral infections. Conventional treatments for such condition fell short of providing satisfying therapeutic results. Recent advances in organ regeneration therapy which utilize tissue stem cells to fabricate bioengineered 3D organ buds, have introduced a promising therapeutic tool for full functional organ regeneration. However, finding a sustainable and easily accessible cell source for such approaches is still challenging, especially in case of severely atrophied tissues such as irradiated salivary glands. In response to this, we hypothesized that bone marrow derived mesenchymal stem cells (MSCs) could be used as feeder cells to induce salivary epithelial tissues/cells branching. Indeed, in 2D cultures, MSCs supported branching of embryonic submandibular salivary gland (SMG) epithelium. Interestingly, this enhancing effect was dependent on the initial number of MSC feeder cells. In addition, MSCs supported the self-assembly of SMG epithelial progenitor cells into well-patterned and branched 3D salivary organoids. Therefore, these findings propose MSCs as a valuable candidate cell source for induced SMG epithelial branching, which can potentially be applied in future methods for SMG regeneration approaches.
Collapse
Affiliation(s)
- Mahmoud Farahat
- Department of Biomaterials, Okayama University, Okayama, Japan
- Department of Oral Rehabilitation and Regenerative Medicine, Okayama University, Okayama, Japan
| | - Gulsan Ara Sathi
- Department of Bio-Systems Engineering, Graduate School of Science and Engineering, Yamagata University, Yamagata, Japan
| | | | - Hiroaki Taketa
- Department of Biomaterials, Okayama University, Okayama, Japan
- Center for the Development of Medical and Health Care Education, Okayama University, Okayama, Japan
| | - Takuo Kuboki
- Department of Oral Rehabilitation and Regenerative Medicine, Okayama University, Okayama, Japan
| | | |
Collapse
|
71
|
Lakshmanan R, Ukani G, Rishi MT, Maulik N. Trimodal rescue of hind limb ischemia with growth factors, cells, and nanocarriers: fundamentals to clinical trials. Can J Physiol Pharmacol 2017; 95:1125-1140. [PMID: 28407473 DOI: 10.1139/cjpp-2016-0713] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Peripheral artery disease is a severe medical condition commonly characterized by critical or acute limb ischemia. Gradual accumulation of thrombotic plaques in peripheral arteries of the lower limb may lead to intermittent claudication or ischemia in muscle tissue. Ischemic muscle tissue with lesions may become infected, resulting in a non-healing wound. Stable progression of the non-healing wound associated with severe ischemia might lead to functional deterioration of the limb, which, depending on the severity, can result in amputation. Immediate rescue of ischemic muscles through revascularization strategies is considered the gold standard to treat critical limb ischemia. Growth factors offer multiple levels of protection in revascularization of ischemic tissue. In this review, the basic mechanism through which growth factors exert their beneficial properties to rescue the ischemic limb is extensively discussed. Moreover, clinical trials based on growth factor and stem cell therapy to treat critical limb ischemia are considered. The clinical utility of stem cell therapy for the treatment of limb ischemia is explained and recent advances in nanocarrier technology for selective growth factor and stem cell supplementation are summarized.
Collapse
Affiliation(s)
- Rajesh Lakshmanan
- Molecular Cardiology and Angiogenesis Laboratory, Department of Surgery, University of Connecticut School of Medicine, Farmington, CT 06030, USA.,Molecular Cardiology and Angiogenesis Laboratory, Department of Surgery, University of Connecticut School of Medicine, Farmington, CT 06030, USA
| | - Gopi Ukani
- Molecular Cardiology and Angiogenesis Laboratory, Department of Surgery, University of Connecticut School of Medicine, Farmington, CT 06030, USA.,Molecular Cardiology and Angiogenesis Laboratory, Department of Surgery, University of Connecticut School of Medicine, Farmington, CT 06030, USA
| | - Muhammad Tipu Rishi
- Molecular Cardiology and Angiogenesis Laboratory, Department of Surgery, University of Connecticut School of Medicine, Farmington, CT 06030, USA.,Molecular Cardiology and Angiogenesis Laboratory, Department of Surgery, University of Connecticut School of Medicine, Farmington, CT 06030, USA
| | - Nilanjana Maulik
- Molecular Cardiology and Angiogenesis Laboratory, Department of Surgery, University of Connecticut School of Medicine, Farmington, CT 06030, USA.,Molecular Cardiology and Angiogenesis Laboratory, Department of Surgery, University of Connecticut School of Medicine, Farmington, CT 06030, USA
| |
Collapse
|
72
|
Liu WT, Jing YY, Yu GF, Chen H, Han ZP, Yu DD, Fan QM, Ye F, Li R, Gao L, Zhao QD, Wu MC, Wei LX. Hepatic stellate cell promoted hepatoma cell invasion via the HGF/c-Met signaling pathway regulated by p53. Cell Cycle 2017; 15:886-94. [PMID: 27077227 DOI: 10.1080/15384101.2016.1152428] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
Abstract
The biological behaviors of hepatocellular carcinoma (HCC) are complex mainly due to heterogeneity of progressive genetic and epigenetic mutations as well as tumor environment. Hepatocyte growth factor (HGF)/c-Met signaling pathway is regarded to be a prototypical example for stromal-epithelial interactions during developmental morphogenesis, wound healing, organ regeneration and cancer progression. And p53 plays as an important regulator of Met-dependent cell motility and invasion. Present study showed that 2 HCC cell lines, Hep3B and HepG2, displayed different invasive capacity when treated with HGF which was secreted by hepatic stellate cells (HSCs). We found that HGF promoted Hep3B cells invasion and migration as well as epithelial-mesenchymal transition (EMT) occurrence because Hep3B was p53 deficient, which leaded to the c-Met over-expression. Then we found that HGF/c-Met promoted Hep3B cells invasion and migration by upregulating Snail expression. In conclusion, HGF/c-Met signaling is enhanced by loss of p53 expression, resulting in increased ability of invasion and migration by upregulating the expression of Snail.
Collapse
Affiliation(s)
- Wen-Ting Liu
- a Tumor Immunology and Gene Therapy Center, Eastern Hepatobiliary Surgery Hospital, The Second Military Medical University , Shanghai , China
| | - Ying-Ying Jing
- a Tumor Immunology and Gene Therapy Center, Eastern Hepatobiliary Surgery Hospital, The Second Military Medical University , Shanghai , China
| | - Guo-feng Yu
- c Oncology Department , Ji'an Hospital, Shanghai East Hospital, Tongji University School of Medicine , Shanghai , China
| | - Hong Chen
- d Pathology Department , Funing Hospital of Traditional Chinese Medicine , Qinghuangdao , Hebei Province , China
| | - Zhi-peng Han
- a Tumor Immunology and Gene Therapy Center, Eastern Hepatobiliary Surgery Hospital, The Second Military Medical University , Shanghai , China
| | - Dan-Dan Yu
- a Tumor Immunology and Gene Therapy Center, Eastern Hepatobiliary Surgery Hospital, The Second Military Medical University , Shanghai , China
| | - Qing-Min Fan
- e Ultrasonography Department , The First Affiliated Hospital of Soochow University , Jiangsu , China
| | - Fei Ye
- a Tumor Immunology and Gene Therapy Center, Eastern Hepatobiliary Surgery Hospital, The Second Military Medical University , Shanghai , China
| | - Rong Li
- a Tumor Immunology and Gene Therapy Center, Eastern Hepatobiliary Surgery Hospital, The Second Military Medical University , Shanghai , China
| | - Lu Gao
- a Tumor Immunology and Gene Therapy Center, Eastern Hepatobiliary Surgery Hospital, The Second Military Medical University , Shanghai , China
| | - Qiu-Dong Zhao
- a Tumor Immunology and Gene Therapy Center, Eastern Hepatobiliary Surgery Hospital, The Second Military Medical University , Shanghai , China
| | - Meng-Chao Wu
- b Department of Comprehensive Treatment , Eastern Hepatobiliary Surgery Hospital, The Second Military Medical University , Shanghai , China
| | - Li-Xin Wei
- a Tumor Immunology and Gene Therapy Center, Eastern Hepatobiliary Surgery Hospital, The Second Military Medical University , Shanghai , China
| |
Collapse
|
73
|
Szturz P, Raymond E, Abitbol C, Albert S, de Gramont A, Faivre S. Understanding c-MET signalling in squamous cell carcinoma of the head & neck. Crit Rev Oncol Hematol 2017; 111:39-51. [DOI: 10.1016/j.critrevonc.2017.01.004] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2016] [Revised: 10/28/2016] [Accepted: 01/09/2017] [Indexed: 12/21/2022] Open
|
74
|
Epithelial-mesenchymal transition in morphogenesis, cancer progression and angiogenesis. Exp Cell Res 2017; 353:1-5. [PMID: 28257786 DOI: 10.1016/j.yexcr.2017.02.041] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2017] [Revised: 02/22/2017] [Accepted: 02/27/2017] [Indexed: 12/18/2022]
Abstract
All organs consist of an epithelium and an associated mesenchyme, so these epithelial-mesenchymal intercations are among the most important phenomena in nature. The aim of this article is the summarize the common mechanisms involved in the establishment of epithelial mesenchymal transition in three biological processes, namely organogenesis, tumor progression and metastasis, and angiogenesis, apparently independent each from other. A common feature of these processes is the fact that specialized epithelial cells lose their features, including cell adhesion and polarity, reorganize their cytoskeleton, and acquire a mesenchymal morphology and the ability to migrate.
Collapse
|
75
|
Hepatocyte growth factor secreted by bone marrow stem cell reduce ER stress and improves repair in alveolar epithelial II cells. Sci Rep 2017; 7:41901. [PMID: 28157203 PMCID: PMC5291222 DOI: 10.1038/srep41901] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Accepted: 01/03/2017] [Indexed: 01/04/2023] Open
Abstract
Idiopathic Pulmonary Fibrosis (IPF) is a progressive, irreversible lung disease with complex pathophysiology. Evidence of endoplasmic reticulum (ER) stress has been reported in alveolar epithelial cells (AEC) in IPF patients. Secreted mediators from bone marrow stem cells (BMSC-cm) have regenerative properties. In this study we investigate the beneficial effects of BMSC-cm on ER stress response in primary AEC and ER stressed A549 cells. We hypothesize that BMSC-cm reduces ER stress. Primary AEC isolated from IPF patients were treated with BMSC-cm. To induce ER stress A549 cells were incubated with Tunicamycin or Thapsigargin and treated with BMSC-cm, or control media. Primary IPF-AEC had high Grp78 and CHOP gene expression, which was lowered after BMSC-cm treatment. Similar results were observed in ER stressed A549 cells. Alveolar epithelial repair increased in presence of BMSC-cm in ER stressed A549 cells. Hepatocyte growth factor (HGF) was detected in biologically relevant levels in BMSC-cm. Neutralization of HGF in BMSC-cm attenuated the beneficial effects of BMSC-cm including synthesis of surfactant protein C (SP-C) in primary AEC, indicating a crucial role of HGF in ER homeostasis and alveolar epithelial repair. Our data suggest that BMSC-cm may be a potential therapeutic option for treating pulmonary fibrosis.
Collapse
|
76
|
Imamura R, Matsumoto K. Hepatocyte growth factor in physiology and infectious diseases. Cytokine 2017; 98:97-106. [PMID: 28094206 DOI: 10.1016/j.cyto.2016.12.025] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Revised: 12/26/2016] [Accepted: 12/26/2016] [Indexed: 01/14/2023]
Abstract
Hepatocyte growth factor (HGF) is a pleiotropic cytokine composed of an α-chain and a β-chain, and these chains contain four kringle domains and a serine protease-like structure, respectively. The receptor for HGF was identified as the c-met proto-oncogene product of transmembrane receptor tyrosine kinase. HGF-induced signaling through the receptor Met provokes dynamic biological responses that support morphogenesis, regeneration, and the survival of various cells and tissues, which includes hepatocytes, renal tubular cells, and neurons. Characterization of tissue-specific Met knockout mice has further indicated that the HGF-Met system modulates immune cell functions and also plays an inhibitory role in the progression of chronic inflammation and fibrosis. However, the biological actions that are driven by the HGF-Met pathway all play a role in the acquisition of the malignant characteristics in tumor cells, such as invasion, metastasis, and drug resistance in the tumor microenvironment. Even though oncogenic Met signaling remains the major research focus, the HGF-Met axis has also been implicated in infectious diseases. Many pathogens try to utilize host HGF-Met system to establish comfortable environment for infection. Their strategies are not only simply change the expression level of HGF or Met, but also actively hijack HGF-Met system and deregulating Met signaling using their pathogenic factors. Consequently, the monitoring of HGF and Met expression, along with real-time detection of Met activation, can be a beneficial biomarker of these infectious diseases. Preclinical studies designed to address the therapeutic significance of HGF have been performed on injury/disease models, including acute tissue injury, chronic fibrosis, and cardiovascular and neurodegenerative diseases. Likewise, manipulating the HGF-Met system with complete control will lead to a tailor made treatment for those infectious diseases.
Collapse
Affiliation(s)
- Ryu Imamura
- Division of Tumor Dynamics and Regulation, Cancer Research Institute, Kanazawa University, Kakuma-machi, Kanazawa 920-1192, Japan
| | - Kunio Matsumoto
- Division of Tumor Dynamics and Regulation, Cancer Research Institute, Kanazawa University, Kakuma-machi, Kanazawa 920-1192, Japan.
| |
Collapse
|
77
|
Senthivel VR, Sturrock M, Piedrafita G, Isalan M. Identifying ultrasensitive HGF dose-response functions in a 3D mammalian system for synthetic morphogenesis. Sci Rep 2016; 6:39178. [PMID: 27982133 PMCID: PMC5159920 DOI: 10.1038/srep39178] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Accepted: 11/18/2016] [Indexed: 02/06/2023] Open
Abstract
Nonlinear responses to signals are widespread natural phenomena that affect various cellular processes. Nonlinearity can be a desirable characteristic for engineering living organisms because it can lead to more switch-like responses, similar to those underlying the wiring in electronics. Steeper functions are described as ultrasensitive, and can be applied in synthetic biology by using various techniques including receptor decoys, multiple co-operative binding sites, and sequential positive feedbacks. Here, we explore the inherent non-linearity of a biological signaling system to identify functions that can potentially be exploited using cell genome engineering. For this, we performed genome-wide transcription profiling to identify genes with ultrasensitive response functions to Hepatocyte Growth Factor (HGF). We identified 3,527 genes that react to increasing concentrations of HGF, in Madin-Darby canine kidney (MDCK) cells, grown as cysts in 3D collagen cell culture. By fitting a generic Hill function to the dose-responses of these genes we obtained a measure of the ultrasensitivity of HGF-responsive genes, identifying a subset with higher apparent Hill coefficients (e.g. MMP1, TIMP1, SNORD75, SNORD86 and ERRFI1). The regulatory regions of these genes are potential candidates for future engineering of synthetic mammalian gene circuits requiring nonlinear responses to HGF signalling.
Collapse
Affiliation(s)
- Vivek Raj Senthivel
- Department of Life Sciences, Imperial College London, London SW7 2AZ, United Kingdom.,EMBL/CRG Systems Biology Research Unit, Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Dr. Aiguader 88, Barcelona 08003, Spain.,Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Marc Sturrock
- Department of Life Sciences, Imperial College London, London SW7 2AZ, United Kingdom
| | - Gabriel Piedrafita
- Department of Life Sciences, Imperial College London, London SW7 2AZ, United Kingdom.,Department of Biochemistry and Cambridge Systems Biology Centre, University of Cambridge, Cambridge CB2 1GA, UK
| | - Mark Isalan
- Department of Life Sciences, Imperial College London, London SW7 2AZ, United Kingdom
| |
Collapse
|
78
|
Liu J, Wang W, Liu M, Su L, Zhou H, Xia Y, Ran J, Lin HY, Yang B. Repulsive guidance molecule b inhibits renal cyst development through the bone morphogenetic protein signaling pathway. Cell Signal 2016; 28:1842-1851. [DOI: 10.1016/j.cellsig.2016.08.015] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Revised: 08/25/2016] [Accepted: 08/25/2016] [Indexed: 02/04/2023]
|
79
|
Lail-Trecker M, Gulati R, Peluso JJ. A Role for Hepatocyte Growth Factors/Scatter Factor in Regulating Normal and Neoplastic Cells of Reproductive Tissues. ACTA ACUST UNITED AC 2016. [DOI: 10.1177/107155769800500302] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Affiliation(s)
| | - Rita Gulati
- Department of Obstetrics and Gynecology, University of Connecticut Health Center, Farmington, Connecticut
| | - John J. Peluso
- Department of Obstetrics and Gynecology, University of Connecticut Health Center, Farmington, CT 06030
| |
Collapse
|
80
|
Lee E, Ewald ML, Sedarous M, Kim T, Weyers BW, Truong RH, Yamada S. Deletion of the cytoplasmic domain of N-cadherin reduces, but does not eliminate, traction force-transmission. Biochem Biophys Res Commun 2016; 478:1640-6. [PMID: 27596967 DOI: 10.1016/j.bbrc.2016.08.173] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Accepted: 08/30/2016] [Indexed: 10/21/2022]
Abstract
Collective migration of epithelial cells is an integral part of embryonic development, wound healing, tissue renewal and carcinoma invasion. While previous studies have focused on cell-extracellular matrix adhesion as a site of migration-driving, traction force-transmission, cadherin mediated cell-cell adhesion is also capable of force-transmission. Using a soft elastomer coated with purified N-cadherin as a substrate and a Hepatocyte Growth Factor-treated, transformed MDCK epithelial cell line as a model system, we quantified traction transmitted by N-cadherin-mediated contacts. On a substrate coated with purified extracellular domain of N-cadherin, cell surface N-cadherin proteins arranged into puncta. N-cadherin mutants (either the cytoplasmic deletion or actin-binding domain chimera), however, failed to assemble into puncta, suggesting the assembly of focal adhesion like puncta requires the cytoplasmic domain of N-cadherin. Furthermore, the cytoplasmic domain deleted N-cadherin expressing cells exerted lower traction stress than the full-length or the actin binding domain chimeric N-cadherin. Our data demonstrate that N-cadherin junctions exert significant traction stress that requires the cytoplasmic domain of N-cadherin, but the loss of the cytoplasmic domain does not completely eliminate traction force transmission.
Collapse
Affiliation(s)
- Eliot Lee
- Biomedical Engineering Department, University of California, Davis, United States
| | - Makena L Ewald
- Biomedical Engineering Department, University of California, Davis, United States
| | - Mary Sedarous
- Biomedical Engineering Department, University of California, Davis, United States
| | - Timothy Kim
- Biomedical Engineering Department, University of California, Davis, United States
| | - Brent W Weyers
- Biomedical Engineering Department, University of California, Davis, United States
| | - Rose Hong Truong
- Biomedical Engineering Department, University of California, Davis, United States
| | - Soichiro Yamada
- Biomedical Engineering Department, University of California, Davis, United States.
| |
Collapse
|
81
|
Ravikrishnan A, Ozdemir T, Bah M, Baskerville KA, Shah SI, Rajasekaran AK, Jia X. Regulation of Epithelial-to-Mesenchymal Transition Using Biomimetic Fibrous Scaffolds. ACS APPLIED MATERIALS & INTERFACES 2016; 8:17915-26. [PMID: 27322677 PMCID: PMC5070665 DOI: 10.1021/acsami.6b05646] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Epithelial-to-mesenchymal transition (EMT) is a well-studied biological process that takes place during embryogenesis, carcinogenesis, and tissue fibrosis. During EMT, the polarized epithelial cells with a cuboidal architecture adopt an elongated fibroblast-like morphology. This process is accompanied by the expression of many EMT-specific molecular markers. Although the molecular mechanism leading to EMT has been well-established, the effects of matrix topography and microstructure have not been clearly elucidated. Synthetic scaffolds mimicking the meshlike structure of the basement membrane with an average fiber diameter of 0.5 and 5 μm were produced via the electrospinning of poly(ε-caprolactone) (PCL) and were used to test the significance of fiber diameter on EMT. Cell-adhesive peptide motifs were conjugated to the fiber surface to facilitate cell attachment. Madin-Darby Canine Kidney (MDCK) cells grown on these substrates showed distinct phenotypes. On 0.5 μm substrates, cells grew as compact colonies with an epithelial phenotype. On 5 μm scaffolds, cells were more individually dispersed and appeared more fibroblastic. Upon the addition of hepatocyte growth factor (HGF), an EMT inducer, cells grown on the 0.5 μm scaffold underwent pronounced scattering, as evidenced by the alteration of cell morphology, localization of focal adhesion complex, weakening of cell-cell adhesion, and up-regulation of mesenchymal markers. In contrast, HGF did not induce a pronounced scattering of MDCK cells cultured on the 5.0 μm scaffold. Collectively, our results show that the alteration of the fiber diameter of proteins found in the basement membrane may create enough disturbances in epithelial organization and scattering that might have important implications in disease progression.
Collapse
Affiliation(s)
- Anitha Ravikrishnan
- Department of Materials Science and Engineering, University of Delaware, Newark, DE 19716, USA
| | - Tugba Ozdemir
- Department of Materials Science and Engineering, University of Delaware, Newark, DE 19716, USA
| | - Mohamed Bah
- Department of Materials Science and Engineering, University of Delaware, Newark, DE 19716, USA
| | | | - S. Ismat Shah
- Department of Materials Science and Engineering, University of Delaware, Newark, DE 19716, USA
- Department of Physics and Astronomy, University of Delaware, Newark, DE 19716, USA
| | - Ayyappan K. Rajasekaran
- Department of Materials Science and Engineering, University of Delaware, Newark, DE 19716, USA
- Department of Biological Sciences, University of Delaware, Newark, DE, 19716, USA
- Therapy Architects, LLC, Helen F Graham Cancer Center, Newark, DE, 19718, USA
| | - Xinqiao Jia
- Department of Materials Science and Engineering, University of Delaware, Newark, DE 19716, USA
- Department of Biological Sciences, University of Delaware, Newark, DE, 19716, USA
- Department of Biomedical Engineering, University of Delaware, Newark, DE 19716, USA
- To whom correspondence should be addressed: Xinqiao Jia, 201 DuPont Hall, Department of Materials Science and Engineering, University of Delaware, Newark, DE, 19716, USA. Phone: 302-831-6553, Fax: 302-831-4545,
| |
Collapse
|
82
|
Yang YM, Lee CG, Koo JH, Kim TH, Lee JM, An J, Kim KM, Kim SG. Gα12 overexpressed in hepatocellular carcinoma reduces microRNA-122 expression via HNF4α inactivation, which causes c-Met induction. Oncotarget 2016; 6:19055-69. [PMID: 25965999 PMCID: PMC4662475 DOI: 10.18632/oncotarget.3957] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Accepted: 04/08/2015] [Indexed: 12/12/2022] Open
Abstract
MicroRNA-122 (miR-122) is implicated as a regulator of physiological and pathophysiological processes in the liver. Overexpression of Gα12 is associated with overall survival in patients with hepatocellular carcinoma (HCC). Array-based miRNA profiling was performed on Huh7 stably transfected with activated Gα12 to find miRNAs regulated by the Gα12 pathway; among them, miR-122 was most greatly repressed. miR-122 directly inhibits c-Met expression, playing a role in HCC progression. Gα12 destabilized HNF4α by accelerating ubiquitination, impeding constitutive expression of miR-122. miR-122 mimic transfection diminished the ability of Gα12 to increase c-Met and to activate ERK, STAT3, and Akt/mTOR, suppressing cell proliferation with augmented apoptosis. Consistently, miR-122 transfection prohibited tumor cell colony formation and endothelial tube formation. In a xenograft model, Gα12 knockdown attenuated c-Met expression by restoring HNF4α levels, and elicited tumor cell apoptosis but diminished Ki67 intensities. In human HCC samples, Gα12 levels correlated to c-Met and were inversely associated with miR-122. Both miR-122 and c-Met expression significantly changed in tumor node metastasis (TNM) stage II/III tumors. Moreover, changes in Gα12 and miR-122 levels discriminated recurrence-free and overall survival rates of HCC patients. Collectively, Gα12 overexpression in HCC inhibits MIR122 transactivation by inactivating HNF4α, which causes c-Met induction, contributing to cancer aggressiveness.
Collapse
Affiliation(s)
- Yoon Mee Yang
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Korea
| | - Chan Gyu Lee
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Korea
| | - Ja Hyun Koo
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Korea
| | - Tae Hyun Kim
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Korea
| | - Jung Min Lee
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Korea
| | - Jihyun An
- Department of Internal Medicine, Asan Liver Center, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Kang Mo Kim
- Department of Internal Medicine, Asan Liver Center, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Sang Geon Kim
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Korea
| |
Collapse
|
83
|
Islam MR, Yamagami K, Yoshii Y, Yamauchi N. Growth factor induced proliferation, migration, and lumen formation of rat endometrial epithelial cells in vitro. J Reprod Dev 2016; 62:271-8. [PMID: 26946922 PMCID: PMC4919291 DOI: 10.1262/jrd.2015-158] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Accepted: 02/13/2016] [Indexed: 12/11/2022] Open
Abstract
Endometrial modulation is essential for the preservation of normal uterine physiology, and this modulation is driven by a number of growth factors. The present study investigated the mitogenic, motogenic, and morphogenic effects of epidermal growth factor (EGF) and hepatocyte growth factor (HGF) on rat endometrial epithelial (REE) cells. The REE cells were isolated and cultured and then characterized based on their morphology and their expression of epithelial cell markers. The MTT assay revealed that EGF and HGF induce proliferation of REE cells. Consistent with increased proliferation, we found that the cell cycle regulatory factor Cyclin D1 was also upregulated upon EGF and HGF addition. REE cell migration was prompted by EGF, as observed with the Oris Cell Migration Assay. The morphogenic impact of growth factors on REE cells was studied in a three-dimensional BD Matrigel cell culture system, wherein these growth factors also increased the frequency of lumen formation. In summary, we show that EGF and HGF have a stimulatory effect on REE cells, promoting proliferation, cell migration, and lumen formation. Our findings provide important insights that further the understanding of endometrial regeneration and its regulation.
Collapse
Affiliation(s)
- Md Rashedul Islam
- Department of Animal and Marine Bioresource Sciences, Faculty of Agriculture, Kyushu University, Fukuoka 812-8581, Japan
| | | | | | | |
Collapse
|
84
|
Ilangumaran S, Villalobos-Hernandez A, Bobbala D, Ramanathan S. The hepatocyte growth factor (HGF)–MET receptor tyrosine kinase signaling pathway: Diverse roles in modulating immune cell functions. Cytokine 2016; 82:125-39. [PMID: 26822708 DOI: 10.1016/j.cyto.2015.12.013] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2015] [Revised: 12/11/2015] [Accepted: 12/12/2015] [Indexed: 12/14/2022]
|
85
|
Haws HJ, McNeil MA, Hansen MDH. Control of cell mechanics by RhoA and calcium fluxes during epithelial scattering. Tissue Barriers 2016; 4:e1187326. [PMID: 27583192 DOI: 10.1080/21688370.2016.1187326] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Revised: 04/25/2016] [Accepted: 05/01/2016] [Indexed: 12/17/2022] Open
Abstract
Epithelial tissues use adherens junctions to maintain tight interactions and coordinate cellular activities. Adherens junctions are remodeled during epithelial morphogenesis, including instances of epithelial-mesenchymal transition, or EMT, wherein individual cells detach from the tissue and migrate as individual cells. EMT has been recapitulated by growth factor induction of epithelial scattering in cell culture. In culture systems, cells undergo a highly reproducible series of cell morphology changes, most notably cell spreading followed by cellular compaction and cell migration. These morphology changes are accompanied by striking actin rearrangements. The current evidence suggests that global changes in actomyosin-based cellular contractility, first a loss of contractility during spreading and its activation during cell compaction, are the main drivers of epithelial scattering. In this review, we focus on how spreading and contractility might be controlled during epithelial scattering. While we propose a central role for RhoA, which is well known to control cellular contractility in multiple systems and whose role in epithelial scattering is well accepted, we suggest potential roles for additional cellular systems whose role in epithelial cell biology has been less well documented. In particular, we propose critical roles for vesicle recycling, calcium channels, and calcium-dependent kinases.
Collapse
Affiliation(s)
- Hillary J Haws
- Physiology and Developmental Biology, Brigham Young University , Provo, UT, USA
| | - Melissa A McNeil
- Physiology and Developmental Biology, Brigham Young University , Provo, UT, USA
| | - Marc D H Hansen
- Physiology and Developmental Biology, Brigham Young University , Provo, UT, USA
| |
Collapse
|
86
|
Kato T, Oka K, Nakamura T, Ito A. Decreased expression of Met during differentiation in rat lung. Eur J Histochem 2016; 60:2575. [PMID: 26972715 PMCID: PMC4800251 DOI: 10.4081/ejh.2016.2575] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2015] [Revised: 01/05/2016] [Accepted: 01/05/2016] [Indexed: 12/02/2022] Open
Abstract
Organ-specific stem cells play key roles in maintaining the epithelial cell layers of lung. Bronchioalveolar stem cells (BASCs) are distal lung epithelial stem cells of adult mice. Alveolar type 2 (AT2) cells have important functions and serve as progenitor cells of alveolar type 1 (AT1) cells to repair the epithelium when they are injured. Hepatocyte growth factor (HGF) elicits mitogenic, morphogenic, and anti-apoptotic effects on lung epithelial cells through tyrosine phosphorylation of Met receptor, and thus is recognized as a pulmotrophic factor. To understand which cells HGF targets in lung, we identified the cells expressing Met by immunofluorescence assay. Met was strongly expressed in BASCs, which expressed an AT2 cell marker, pro-SP-C, and a club cell marker, CCSP. In alveoli, we found higher expression of Met in primary AT2 than in AT1 cells, which was confirmed using primary AT2 cells. We further examined the mitogenic activity of HGF in AT2-cell-derived alveolar-like cysts (ALCs) in 3D culture. Multicellular ALCs expressed Met, and HGF enhanced the ALC production. Taking these findings together, BASCs could also be an important target for HGF, and HGF-Met signaling could function more potent on cells that have greater multipotency in adult lung.
Collapse
|
87
|
Matouk IJ, Halle D, Raveh E, Gilon M, Sorin V, Hochberg A. The role of the oncofetal H19 lncRNA in tumor metastasis: orchestrating the EMT-MET decision. Oncotarget 2016; 7:3748-65. [PMID: 26623562 PMCID: PMC4826167 DOI: 10.18632/oncotarget.6387] [Citation(s) in RCA: 107] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Accepted: 11/15/2015] [Indexed: 12/14/2022] Open
Abstract
Long non-coding RNA (lncRNA) genes are emerging as key players in the metastatic cascade. Current evidence indicate that H19 lncRNA and the microRNA(miRNA) miR-675, which is processed from it, play crucial roles in metastasis, through the regulation of critical events specifically the epithelial to mesenchymal (EMT) and the mesenchymal to epithelial transitions (MET). This review summarizes recent mechanistic pathways and tries to put together seemingly conflicting data from different reports under one proposed general scheme underlying the various roles of H19/miR-675 in the metastatic cascade. We propose several approaches to harnessing this knowledge for translational medicine.
Collapse
Affiliation(s)
- Imad J. Matouk
- Department of Biological Chemistry, Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel
- Department of Biological Sciences, Faculty of Science and Technology, Al-Quds University, Jerusalem, West Bank
| | - David Halle
- Department of Biological Chemistry, Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Eli Raveh
- Department of Biological Chemistry, Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Michal Gilon
- Department of Biological Chemistry, Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Vladimir Sorin
- Department of Biological Chemistry, Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Avraham Hochberg
- Department of Biological Chemistry, Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel
| |
Collapse
|
88
|
Rioja AY, Tiruvannamalai Annamalai R, Paris S, Putnam AJ, Stegemann JP. Endothelial sprouting and network formation in collagen- and fibrin-based modular microbeads. Acta Biomater 2016; 29:33-41. [PMID: 26481042 PMCID: PMC4681647 DOI: 10.1016/j.actbio.2015.10.022] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Revised: 09/04/2015] [Accepted: 10/15/2015] [Indexed: 12/18/2022]
Abstract
A modular tissue engineering approach may have advantages over current therapies in providing rapid and sustained revascularization of ischemic tissue. In this study, modular protein microbeads were prepared from pure fibrin (FIB) and collagen-fibrin composites (COL-FIB) using a simple water-in-oil emulsification technique. Human endothelial cells and fibroblasts were embedded directly in the microbead matrix. The resulting microbeads were generally spheroidal with a diameter of 100-200μm. Cell viability was high (75-80% viable) in microbeads, but was marginally lower than in bulk hydrogels of corresponding composition (85-90% viable). Cell proliferation was significantly greater in COL-FIB microbeads after two weeks in culture, compared to pure FIB microbeads. Upon embedding of microbeads in a surrounding fibrin hydrogel, endothelial cell networks formed inside the microbead matrix and extended into the surrounding matrix. The number of vessel segments, average segment length, and number of branch points was higher in FIB samples, compared to COL-FIB samples, resulting in significantly longer total vessel networks. Anastomosis of vessel networks from adjacent microbeads was also observed. These studies demonstrate that primitive vessel networks can be formed by modular protein microbeads containing embedded endothelial cells and fibroblasts. Such microbeads may find utility as prevascularized tissue modules that can be delivered minimally invasively as a therapy to restore blood flow to ischemic tissues. STATEMENT OF SIGNIFICANCE Vascularization is critically important for tissue engineering and regenerative medicine, and materials that support and/or promote neovascularization are of value both for translational applications and for mechanistic studies and discovery-based research. Therefore, we fabricated small modular microbeads formulated from pure fibrin (FIB) and collagen-fibrin (COL-FIB) containing endothelial cells and supportive fibroblasts. We explored how cells encapsulated within these materials form microvessel-like networks both within and outside of the microbeads when embedded in larger 3D matrices. FIB microbeads were found to initiate more extensive sprouting into the surrounding ECM in vitro. These results represent an important step towards our goal of developing injectable biomaterial modules containing preformed vascular units that can rapidly restore vascularization to an ischemic tissue in vivo.
Collapse
Affiliation(s)
- Ana Y Rioja
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48105, United States
| | | | - Spencer Paris
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48105, United States
| | - Andrew J Putnam
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48105, United States.
| | - Jan P Stegemann
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48105, United States.
| |
Collapse
|
89
|
Kato T, Oka K, Nakamura T, Ito A. Bronchioalveolar morphogenesis of human bronchial epithelial cells depending upon hepatocyte growth factor. J Cell Mol Med 2015; 19:2818-26. [PMID: 26416301 PMCID: PMC4687712 DOI: 10.1111/jcmm.12672] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2014] [Accepted: 07/22/2015] [Indexed: 01/09/2023] Open
Abstract
Lung alveolar regeneration occurs in adult human lungs as a result of proliferation, differentiation and alveolar morphogenesis of stem cells. It is increasingly being believed that bronchial epithelial cells (BECs) have a potential as stem cells, because they are potent to differentiate into multiple central and peripheral lung cell types in three‐dimensional (3D) cultures, and they develop multiple foci with well‐differentiated histogenesis after transformed into neoplastic cells. In this study, we investigated morphogenic abilities of HBE135 human BECs immortalized by E6/E7 oncogene in 3D cultures. When HBE135 cells were cultured alone or co‐cultured with endothelial cells, the cells formed spherical colonies without branching. However, in co‐culture with lung fibroblast MRC‐9 cells, HBE135 cells formed colonies with bronchioalveolar‐like complex branching, suggesting that MRC‐9‐derived soluble factor(s) are responsible for the branching formation. MRC‐9 cells, not endothelial cells, were found to highly express hepatocyte growth factor (HGF), a soluble molecule involved in liver and kidney regeneration. An anti‐HGF neutralizing antibody severely suppressed the complex branching formation, but addition of HGF could not sufficiently compensate the morphogenic effects of MRC‐9 cells, suggesting that MCR‐9‐derived HGF was necessary but insufficient for the bronchioalveolar structure formation. Immunohistochemistry revealed that Met, a cognate receptor for HGF, was highly expressed and phosphorylated in neoplastic BECs from lung adenocarcinomas with well‐differentiated, not poorly differentiated, histogenesis. These results are consistent with the notion that BECs have an aspect of stem cells. This aspect appears to become manifest through HGF–Met signalling pathway activation.
Collapse
Affiliation(s)
- Takashi Kato
- Department of Pathology, Faculty of Medicine, Kinki University, Osaka-Sayama, Osaka, Japan.,Department of Pharmacology, Faculty of Medicine, Kinki University, Osaka-Sayama, Osaka, Japan
| | - Kiyomasa Oka
- Research & Development, Neurogen Inc., Ibaraki, Osaka, Japan
| | | | - Akihiko Ito
- Department of Pathology, Faculty of Medicine, Kinki University, Osaka-Sayama, Osaka, Japan
| |
Collapse
|
90
|
Hepatocyte growth factor in cerebrospinal fluid differentiates community-acquired or nosocomial septic meningitis from other causes of pleocytosis. Fluids Barriers CNS 2015; 12:22. [PMID: 26408034 PMCID: PMC4582940 DOI: 10.1186/s12987-015-0020-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Accepted: 09/16/2015] [Indexed: 01/19/2023] Open
Abstract
Background Due to anatomical restrictions, the inflammatory response to intracerebral bacterial infections exposes swollen brain tissues to pressure and ischemia, resulting in life-threatening damage. Rapid diagnosis and immediate empirical antibiotic therapy is highly important. However, diagnosing meningitis in patients after neurosurgery is complicated, due to brain tissue damage and changes in cerebrospinal fluid (CSF) caused by surgery. Hepatocyte growth factor (HGF) is a local, acute-phase protein with healing properties. Previous studies on community-acquired septic meningitis reported high levels of intrathecally produced HGF. The present study focused on nosocomial meningitis in assessing the levels of HGF in the CSF. Methods HGF concentrations (ELISA) and HGF binding to receptors; c-Met receptor and heparan sulfate proteoglycan were determined in CSF samples (surface plasmon resonance). CSF samples from patients with community-acquired or nosocomial meningitis (217 samples from 135 patients) were compared to those from controls without signs of cerebral nervous system involvement (N = 36) and patients with Alzheimer’s disease (N = 20). Results Compared to samples from patients that had undergone neurosurgery and had other infectious diseases, CSF samples from patients with nosocomial meningitis had significantly higher HGF concentrations (p < 0.001) and binding affinity to c-Met (p < 0.001) and HSPG (p = 0.043) receptors. The sensitivity and specificity to identify nosocomial septic meningitis were 69.7 and 93.4 %, respectively. The HGF concentration and binding affinity to HGF receptors were significantly higher in CSF from patients with community-acquired septic meningitis compared to patients with aseptic (viral and subacute) meningitis as well as controls (p < 0.001). The sensitivity and specificity to identify community-acquired septic meningitis were 95.4 and 95.7 %, respectively. Discussion In febrile nosocomial infections that occurred post neurosurgery, HGF assessment could substantially improve the differentiation of meningitis from other infections and therefore might be a tool for rapid diagnosis, limiting injuries and guiding antibiotic therapy. Electronic supplementary material The online version of this article (doi:10.1186/s12987-015-0020-z) contains supplementary material, which is available to authorized users.
Collapse
|
91
|
Furusawa K, Mizutani T, Machino H, Yahata S, Fukui A, Sasaki N. Application of Multichannel Collagen Gels in Construction of Epithelial Lumen-like Engineered Tissues. ACS Biomater Sci Eng 2015; 1:539-548. [DOI: 10.1021/acsbiomaterials.5b00003] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Kazuya Furusawa
- Faculty of Advanced Life Science, and ‡Division of Biological Sciences
(Macromolecular Functions), School of Science, Hokkaido University, Kita-ku Kita 10 Nishi 8, Sapporo, Hokkaido Japan
| | - Takeomi Mizutani
- Faculty of Advanced Life Science, and ‡Division of Biological Sciences
(Macromolecular Functions), School of Science, Hokkaido University, Kita-ku Kita 10 Nishi 8, Sapporo, Hokkaido Japan
| | - Hiromi Machino
- Faculty of Advanced Life Science, and ‡Division of Biological Sciences
(Macromolecular Functions), School of Science, Hokkaido University, Kita-ku Kita 10 Nishi 8, Sapporo, Hokkaido Japan
| | - Saki Yahata
- Faculty of Advanced Life Science, and ‡Division of Biological Sciences
(Macromolecular Functions), School of Science, Hokkaido University, Kita-ku Kita 10 Nishi 8, Sapporo, Hokkaido Japan
| | - Akimasa Fukui
- Faculty of Advanced Life Science, and ‡Division of Biological Sciences
(Macromolecular Functions), School of Science, Hokkaido University, Kita-ku Kita 10 Nishi 8, Sapporo, Hokkaido Japan
| | - Naoki Sasaki
- Faculty of Advanced Life Science, and ‡Division of Biological Sciences
(Macromolecular Functions), School of Science, Hokkaido University, Kita-ku Kita 10 Nishi 8, Sapporo, Hokkaido Japan
| |
Collapse
|
92
|
Tanimoto A, Yamada T, Nanjo S, Takeuchi S, Ebi H, Kita K, Matsumoto K, Yano S. Receptor ligand-triggered resistance to alectinib and its circumvention by Hsp90 inhibition in EML4-ALK lung cancer cells. Oncotarget 2015; 5:4920-8. [PMID: 24952482 PMCID: PMC4148110 DOI: 10.18632/oncotarget.2055] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Alectinib is a new generation ALK inhibitor with activity against the gatekeeper L1196M mutation that showed remarkable activity in a phase I/II study with echinoderm microtubule associated protein-like 4 (EML4)--anaplastic lymphoma kinase (ALK) non-small cell lung cancer (NSCLC) patients. However, alectinib resistance may eventually develop. Here, we found that EGFR ligands and HGF, a ligand of the MET receptor, activate EGFR and MET, respectively, as alternative pathways, and thereby induce resistance to alectinib. Additionally, the heat shock protein 90 (Hsp90) inhibitor suppressed protein expression of ALK, MET, EGFR, and AKT, and thereby induced apoptosis in EML4-ALK NSCLC cells, even in the presence of EGFR ligands or HGF. These results suggest that Hsp90 inhibitors may overcome ligand-triggered resistance to new generation ALK inhibitors and may result in more successful treatment of NSCLC patients with EML4-ALK.
Collapse
Affiliation(s)
- Azusa Tanimoto
- Divisions of Medical Oncology, Cancer Research Institute, Kanazawa University, Kanazawa, Japan
| | | | | | | | | | | | | | - Seiji Yano
- Divisions of Medical Oncology, Cancer Research Institute, Kanazawa University, Kanazawa, Japan
| |
Collapse
|
93
|
Yevick HG, Duclos G, Bonnet I, Silberzan P. Architecture and migration of an epithelium on a cylindrical wire. Proc Natl Acad Sci U S A 2015; 112:5944-9. [PMID: 25922533 PMCID: PMC4434757 DOI: 10.1073/pnas.1418857112] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
In a wide range of epithelial tissues such as kidney tubules or breast acini, cells organize into bidimensional monolayers experiencing an out-of-plane curvature. Cancer cells can also migrate collectively from epithelial tumors by wrapping around vessels or muscle fibers. However, in vitro experiments dealing with epithelia are mostly performed on flat substrates, neglecting this out-of-plane component. In this paper, we study the development and migration of epithelial tissues on glass wires of well-defined radii varying from less than 1 µm up to 85 µm. To uncouple the effect of out-of-plane curvature from the lateral confinement experienced by the cells in these geometries, we compare our results to experiments performed on narrow adhesive tracks. Because of lateral confinement, the velocity of collective migration increases for radii smaller than typically 20 µm. The monolayer dynamics is then controlled by front-edge protrusions. Conversely, high curvature is identified as the inducer of frequent cell detachments at the front edge, a phenotype reminiscent of the Epithelial-Mesenchymal Transition. High curvature also induces a circumferential alignment of the actin cytoskeleton, stabilized by multiple focal adhesions. This organization of the cytoskeleton is reminiscent of in vivo situations such as the development of the trachea of the Drosophila embryo. Finally, submicron radii halt the monolayer, which then reconfigures into hollow cysts.
Collapse
Affiliation(s)
- Hannah G Yevick
- Laboratoire PhysicoChimie Curie, Institut Curie - Centre de Recherche - Paris Sciences et Lettres, Centre National de la Recherche Scientifique, Université Pierre et Marie Curie - Sorbonne Universités, Equipe labellisée Ligue Contre le Cancer, 75248 Paris, France
| | - Guillaume Duclos
- Laboratoire PhysicoChimie Curie, Institut Curie - Centre de Recherche - Paris Sciences et Lettres, Centre National de la Recherche Scientifique, Université Pierre et Marie Curie - Sorbonne Universités, Equipe labellisée Ligue Contre le Cancer, 75248 Paris, France
| | - Isabelle Bonnet
- Laboratoire PhysicoChimie Curie, Institut Curie - Centre de Recherche - Paris Sciences et Lettres, Centre National de la Recherche Scientifique, Université Pierre et Marie Curie - Sorbonne Universités, Equipe labellisée Ligue Contre le Cancer, 75248 Paris, France
| | - Pascal Silberzan
- Laboratoire PhysicoChimie Curie, Institut Curie - Centre de Recherche - Paris Sciences et Lettres, Centre National de la Recherche Scientifique, Université Pierre et Marie Curie - Sorbonne Universités, Equipe labellisée Ligue Contre le Cancer, 75248 Paris, France
| |
Collapse
|
94
|
Increased c-Met phosphorylation is related to keloid pathogenesis: implications for the biological behaviour of keloid fibroblasts. Pathology 2015; 46:25-31. [PMID: 24300717 DOI: 10.1097/pat.0000000000000028] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Keloid is induced by a pathological wound healing response, and hepatocyte growth factor (HGF) is known to be involved in tissue repair via the activation of its primary receptor, c-Met. We aimed to investigate whether c-Met activation is implicated in keloid pathogenesis. HGF, c-Met, phosphorylated c-Met (p-Met), Ki-67, collagen I protein, and MET gene expression were detected in five normal skin and 30 keloid tissues by immunohistochemistry and quantitative real-time polymerase chain reaction analysis, respectively. The influence of p-Met expression on the biological behaviour of keloid fibroblasts was further investigated with regard to cell proliferation, motility, invasiveness, collagen I expression, and intracellular signaling in vitro. p-Met protein and MET gene expression but not HGF or c-Met protein expression showed significant increases in keloid tissues than dermal layer of normal skin tissues. In keloid tissues, p-Met expression was significantly associated with keloid size, Ki-67 and collagen I expression. Moreover, p-Met expression was also related to proliferation, migration, invasiveness, collagen I expression and activation of AKT and Erk in keloid fibroblasts in vitro. c-Met activation may have a strong influence on keloid pathogenesis, and it can be investigated further as a potential molecular target for keloid therapy.
Collapse
|
95
|
Akita N, Ma N, Okamoto T, Asanuma K, Yoshida K, Nishioka J, Shimaoka M, Suzuki K, Hayashi T. Host protein C inhibitor inhibits tumor growth, but promotes tumor metastasis, which is closely correlated with hypercoagulability. Thromb Res 2015; 135:1203-8. [PMID: 25887633 DOI: 10.1016/j.thromres.2015.03.026] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2014] [Revised: 03/20/2015] [Accepted: 03/30/2015] [Indexed: 10/23/2022]
Abstract
INTRODUCTION Protein C inhibitor (PCI), a member of the serine protease inhibitor family, is expressed in various human tissues, including liver and kidneys. In the plasma, PCI physiologically inhibits an anticoagulant serine protease, activated protein C (APC). PCI expressed by cancer cells suppresses tumor invasion by inhibiting urokinase-type plasminogen activator, and inhibits tumor growth and metastasis, which are independent of its protease-inhibitory activity. In the present study, we clarified the effects of host PCI on growth and metastasis of B16 melanoma (B16) cells by comparing between wild-type mice and mice transgenic for human PCI gene (hPCI-TG), which have a tissue distribution of PCI similar to that observed in humans. MATERIALS AND METHODS Growth of intracutaneously-injected B16 cells was evaluated by measuring the tumor volume, and metastatic behavior of intravenously-injected B16 cells by counting the number of metastatic lung nodules. RESULTS Growth of intracutaneously injected B16 cells was significantly faster in wild-type mice than in hPCI-TG mice; however, hPCI-TG mice developed more metastatic nodules of B16 cells in the lungs. Immunohistochemical analysis using anti-mouse fibrinogen antibody revealed more fibrin deposition in the lung in hPCI-TG mice than in wild-type mice. Furthermore, the more invasive behavior observed in hPCI-TG mice was reduced by rabbit anti-human PCI IgG, APC, or soluble TM administration for 3 consecutive days including the day that B16 cells were injected. CONCLUSIONS Our results suggest that like PCI expressed in tumor cells, host PCI also inhibits tumor growth, but host PCI promotes tumor metastasis via its procoagulant properties.
Collapse
Affiliation(s)
- Nobuyuki Akita
- Faculty of Medical Engineering, Suzuka University of Medical Science, Suzuka-city, Mie, Japan
| | - Ning Ma
- Faculty of Health Science, Suzuka University of Medical Science, Suzuka-city, Mie, Japan
| | - Takayuki Okamoto
- Department of Molecular Pathobiology and Cell Adhesion Biology, Mie University Graduate School of Medicine, Tsu-city, Mie, Japan
| | - Kunihiro Asanuma
- Department of Orthopedic Surgery, Mie University Graduate School of Medicine, Tsu-city, Mie, Japan
| | - Kakunoshin Yoshida
- Department of Orthopedic Surgery, Mie University Graduate School of Medicine, Tsu-city, Mie, Japan
| | - Junji Nishioka
- Department of Clinical Nutrition, Suzuka University of Medical Science, Suzuka-city, Mie, Japan
| | - Motomu Shimaoka
- Department of Molecular Pathobiology and Cell Adhesion Biology, Mie University Graduate School of Medicine, Tsu-city, Mie, Japan
| | - Koji Suzuki
- Faculty of Pharmaceutical Science, Suzuka University of Medical Science, Suzuka-city, Mie, Japan
| | - Tatsuya Hayashi
- Department of Biochemistry, Mie Prefectural College of Nursing, Tsu-city, Mie, Japan.
| |
Collapse
|
96
|
Menis J, Giaj Levra M, Novello S. MET inhibition in lung cancer. Transl Lung Cancer Res 2015; 2:23-39. [PMID: 25806202 DOI: 10.3978/j.issn.2218-6751.2012.12.04] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2012] [Accepted: 12/17/2012] [Indexed: 01/06/2023]
Abstract
Targeted agents have completely changed cancer treatment strategy, leading it from a "one size fits all" approach to a customized therapy. In this scenario Met, a heterodimere receptor tyrosine kinase deeply involved into embryogenesis and organogenesis, has been introduced many years ago as a potential target for biological agents, becoming "druggable" only in this last period of time. Met can be altered through receptor overexpression, genomic amplification, mutations or alternative splicing, autocrine or paracrine secretion of hepatic growth factor (HGF): these dysregulations stimulate tumorigenesis (in terms of cell-cell detachment, proliferation, invasion, angiogenesis and survival) and metastatization. Met is overexpressed in lung cancer and Met gene amplification can drive the dependency of cell survival and proliferation upon the Met signaling. Both Met overexpression and amplification seem to correlate with poor prognosis. Met amplification is also described to be linked to EGFR acquired resistance. Several Met inhibitors have been tested both in preclinical and human trials, demonstrating activity in lung cancer treatment. This paper aims to summarize data on Met biological function, on its interaction with cell signaling and other pathways and to present data on those Met inhibitors currently under evaluation.
Collapse
Affiliation(s)
- Jessica Menis
- Department of Oncology, A.O.U Santa Maria della Misericordia of Udine, Italy
| | - Matteo Giaj Levra
- Department of Oncology, A.O.U San Luigi - Orbassano, University of Turin, Italy
| | - Silvia Novello
- Department of Oncology, A.O.U San Luigi - Orbassano, University of Turin, Italy
| |
Collapse
|
97
|
Sakai K, Aoki S, Matsumoto K. Hepatocyte growth factor and Met in drug discovery. J Biochem 2015; 157:271-84. [PMID: 25770121 DOI: 10.1093/jb/mvv027] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Accepted: 01/13/2015] [Indexed: 12/14/2022] Open
Abstract
Activation of the hepatocyte growth factor (HGF)-Met pathway evokes dynamic biological responses that support the morphogenesis, regeneration and survival of cells and tissues. A characterization of conditional Met knockout mice indicates that the HGF-Met pathway plays important roles in the regeneration, protection and homeostasis of cells such as hepatocytes, renal tubular cells and neurons. Preclinical studies in disease models have indicated that recombinant HGF protein and expression plasmid for HGF are biological drug candidates for the treatment of patients with diseases or injuries that involve impaired tissue function. The phase-I and phase-I/II clinical trials of the intrathecal administration of HGF protein for the treatment of patients with amyotrophic lateral sclerosis and spinal cord injury, respectively, are ongoing. Biological actions of HGF that promote the dynamic movement, morphogenesis and survival of cells also closely participate in invasion-metastasis and resistance to the molecular-targeted drugs in tumour cells. Different types of HGF-Met pathway inhibitors are now in clinical trials for treatment of malignant tumours. Basic research on HGF and Met has lead to drug discoveries in regenerative medicine and tumour biology.
Collapse
Affiliation(s)
- Katsuya Sakai
- Division of Tumor Dynamics and Regulation, Cancer Research Institute, Kanazawa University, Kakuma-machi, Kanazawa 920-1192, Japan; and Department of Bioscience and Bioinformatics, Graduate School of Computer Science and Systems Engineering, Kyushu Institute of Technology, 680-4 Kawazu, Iizuka-shi, Fukuoka 820-8502, Japan
| | - Shunsuke Aoki
- Division of Tumor Dynamics and Regulation, Cancer Research Institute, Kanazawa University, Kakuma-machi, Kanazawa 920-1192, Japan; and Department of Bioscience and Bioinformatics, Graduate School of Computer Science and Systems Engineering, Kyushu Institute of Technology, 680-4 Kawazu, Iizuka-shi, Fukuoka 820-8502, Japan
| | - Kunio Matsumoto
- Division of Tumor Dynamics and Regulation, Cancer Research Institute, Kanazawa University, Kakuma-machi, Kanazawa 920-1192, Japan; and Department of Bioscience and Bioinformatics, Graduate School of Computer Science and Systems Engineering, Kyushu Institute of Technology, 680-4 Kawazu, Iizuka-shi, Fukuoka 820-8502, Japan
| |
Collapse
|
98
|
Ito K, Sakai K, Suzuki Y, Ozawa N, Hatta T, Natsume T, Matsumoto K, Suga H. Artificial human Met agonists based on macrocycle scaffolds. Nat Commun 2015; 6:6373. [PMID: 25758345 PMCID: PMC4382702 DOI: 10.1038/ncomms7373] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2014] [Accepted: 01/23/2015] [Indexed: 12/27/2022] Open
Abstract
Hepatocyte growth factor (HGF) receptor, also known as Met, is a member of the receptor tyrosine kinase family. The Met-HGF interaction regulates various signalling pathways involving downstream kinases, such as Akt and Erk. Met activation is implicated in wound healing of tissues via multiple biological responses triggered by the above-mentioned signalling cascade. Here we report the development of artificial Met-activating dimeric macrocycles. We identify Met-binding monomeric macrocyclic peptides by means of the RaPID (random non-standard peptide integrated discovery) system, and dimerize the respective monomers through rational design. These dimeric macrocycles specifically and strongly activate Met signalling pathways through receptor dimerization and induce various HGF-like cellular responses, such as branching morphogenesis, in human cells. This work suggests our approach for generating dimeric macrocycles as non-protein ligands for cell surface receptors can be useful for developing potential therapeutics with a broad range of potential applications.
Collapse
Affiliation(s)
- Kenichiro Ito
- Department of Chemistry, Graduate School of Science, The University of Tokyo, Tokyo 113-0033, Japan
| | - Katsuya Sakai
- Division of Tumor Dynamics and Regulation, Cancer Research Institute, Kanazawa University, Kanazawa 920-1192, Japan
| | - Yoshinori Suzuki
- Division of Tumor Dynamics and Regulation, Cancer Research Institute, Kanazawa University, Kanazawa 920-1192, Japan
| | - Naoya Ozawa
- Department of Chemistry, Graduate School of Science, The University of Tokyo, Tokyo 113-0033, Japan
| | - Tomohisa Hatta
- National Institute of Advanced Industrial Science and Technology, Biological Information Research Center, Tokyo 135-0064, Japan
| | - Tohru Natsume
- National Institute of Advanced Industrial Science and Technology, Biological Information Research Center, Tokyo 135-0064, Japan
| | - Kunio Matsumoto
- Division of Tumor Dynamics and Regulation, Cancer Research Institute, Kanazawa University, Kanazawa 920-1192, Japan
| | - Hiroaki Suga
- Department of Chemistry, Graduate School of Science, The University of Tokyo, Tokyo 113-0033, Japan
| |
Collapse
|
99
|
Simonneau C, Bérénice Leclercq, Mougel A, Adriaenssens E, Paquet C, Raibaut L, Ollivier N, Drobecq H, Marcoux J, Cianférani S, Tulasne D, de Jonge H, Melnyk O, Vicogne J. Semi-synthesis of a HGF/SF kringle one (K1) domain scaffold generates a potent in vivo MET receptor agonist. Chem Sci 2015; 6:2110-2121. [PMID: 28717459 PMCID: PMC5496502 DOI: 10.1039/c4sc03856h] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Accepted: 01/19/2015] [Indexed: 12/21/2022] Open
Abstract
The development of MET receptor agonists is an important goal in regenerative medicine, but is limited by the complexity and incomplete understanding of its interaction with HGF/SF (Hepatocyte Growth Factor/Scatter Factor). NK1 is a natural occurring agonist comprising the N-terminal (N) and the first kringle (K1) domains of HGF/SF. In the presence of heparin, NK1 can self-associate into a "head to tail" dimer which is considered as the minimal structural module able to trigger MET dimerization and activation whereas isolated K1 and N domains showed a weak or a complete lack of agonistic activity respectively. Starting from these structural and biological observations, we investigated whether it was possible to recapitulate the biological properties of NK1 using a new molecular architecture of isolated N or K1 domains. Therefore, we engineered multivalent N or K1 scaffolds by combining synthetic and homogeneous site-specifically biotinylated N and K1 domains (NB and K1B) and streptavidin (S). NB alone or in complex failed to activate MET signaling and to trigger cellular phenotypes. Importantly and to the contrary of K1B alone, the semi-synthetic K1B/S complex mimicked NK1 MET agonist activity in cell scattering, morphogenesis and survival phenotypic assays. Impressively, K1B/S complex stimulated in vivo angiogenesis and, when injected in mice, protected the liver against fulminant hepatitis in a MET dependent manner whereas NK1 and HGF were substantially less potent. These data reveal that without N domain, proper multimerization of K1 domain is a promising strategy for the rational design of powerful MET agonists.
Collapse
Affiliation(s)
- Claire Simonneau
- UMR CNRS 8161 CNRS , Université de Lille , Institut Pasteur de Lille , 1 rue du Pr Calmette , 59021 Lille Cedex , France . ;
| | - Bérénice Leclercq
- UMR CNRS 8161 CNRS , Université de Lille , Institut Pasteur de Lille , 1 rue du Pr Calmette , 59021 Lille Cedex , France . ;
| | - Alexandra Mougel
- UMR CNRS 8161 CNRS , Université de Lille , Institut Pasteur de Lille , 1 rue du Pr Calmette , 59021 Lille Cedex , France . ;
| | - Eric Adriaenssens
- UMR CNRS 8161 CNRS , Université de Lille , Institut Pasteur de Lille , 1 rue du Pr Calmette , 59021 Lille Cedex , France . ;
| | - Charlotte Paquet
- SIRIC ONCOLille , Maison Régionale de la Recherche Clinique , 6 rue du Pr. Laguesse , 59037 Lille Cedex , France
| | - Laurent Raibaut
- UMR CNRS 8161 CNRS , Université de Lille , Institut Pasteur de Lille , 1 rue du Pr Calmette , 59021 Lille Cedex , France . ;
| | - Nathalie Ollivier
- UMR CNRS 8161 CNRS , Université de Lille , Institut Pasteur de Lille , 1 rue du Pr Calmette , 59021 Lille Cedex , France . ;
| | - Hervé Drobecq
- UMR CNRS 8161 CNRS , Université de Lille , Institut Pasteur de Lille , 1 rue du Pr Calmette , 59021 Lille Cedex , France . ;
| | - Julien Marcoux
- UMR 7178 CNRS , Laboratoire de Spectrométrie de Masse BioOrganique (LSMBO) , IPHC-DSA , Université de Strasbourg , 25 rue Becquerel , 67087 Strasbourg , France
| | - Sarah Cianférani
- UMR 7178 CNRS , Laboratoire de Spectrométrie de Masse BioOrganique (LSMBO) , IPHC-DSA , Université de Strasbourg , 25 rue Becquerel , 67087 Strasbourg , France
| | - David Tulasne
- UMR CNRS 8161 CNRS , Université de Lille , Institut Pasteur de Lille , 1 rue du Pr Calmette , 59021 Lille Cedex , France . ;
| | - Hugo de Jonge
- Division of Immunology and General Pathology , Department of Molecular Medicine , University of Pavia , 9 via A Ferrata , 27100 Pavia , Italy
| | - Oleg Melnyk
- UMR CNRS 8161 CNRS , Université de Lille , Institut Pasteur de Lille , 1 rue du Pr Calmette , 59021 Lille Cedex , France . ;
| | - Jérôme Vicogne
- UMR CNRS 8161 CNRS , Université de Lille , Institut Pasteur de Lille , 1 rue du Pr Calmette , 59021 Lille Cedex , France . ;
| |
Collapse
|
100
|
Katano T, Ootani A, Mizoshita T, Tanida S, Tsukamoto H, Ozeki K, Kataoka H, Joh T. Gastric Mesenchymal Myofibroblasts Maintain Stem Cell Activity and Proliferation of Murine Gastric Epithelium in Vitro. THE AMERICAN JOURNAL OF PATHOLOGY 2015; 185:798-807. [DOI: 10.1016/j.ajpath.2014.11.007] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/08/2014] [Revised: 10/17/2014] [Accepted: 11/07/2014] [Indexed: 01/09/2023]
|