1
|
Escoto A, Hecksel R, Parkinson C, Crane S, Atwell B, King S, Ortiz Chavez D, Jannuzi A, Sands B, Bitler BG, Fehniger TA, Paek AL, Padi M, Schroeder J. Nuclear EGFR in breast cancer suppresses NK cell recruitment and cytotoxicity. Oncogene 2024:10.1038/s41388-024-03211-0. [PMID: 39521886 DOI: 10.1038/s41388-024-03211-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 10/23/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024]
Abstract
Natural Killer (NK) cells can target and destroy cancer cells, yet tumor microenvironments typically suppress NK cell recruitment and cytotoxicity. The epidermal growth factor receptor (EGFR) is a potent oncogene that can activate survival, migration, and proliferation pathways, and clinical data suggests it may also play an immunomodulating role in cancers. Recent work has demonstrated a novel role for nuclear EGFR (nEGFR) in regulating transcriptional events unique from the kinase domain. Using a novel peptide therapeutic (cSNX1.3) that inhibits retrograde trafficking of EGFR and an EGFR nuclear localization mutant, we discovered that nEGFR suppresses NK cell recruitment and cytotoxicity. RNA-Seq analysis of breast cancer cells treated with cSNX1.3 or modified to lack a nuclear localization sequence (EGFRΔNLS) revealed the EGF-dependent induction of NK activating receptor ligands, while kinase inhibition by erlotinib did not impact these genes. NanoString analysis of tumor-bearing WAP-TGFα transgenic mice treated with cSNX1.3 demonstrated an increase in immune cell populations and activating genes. Additionally, immunohistochemistry confirmed an increase in NK cells upon cSNX1.3 treatment. Finally, cSNX1.3 treatment was found to enhance NK cell recruitment and cytotoxicity in vitro. Together, the data demonstrate a unique immunomodulatory role for nEGFR.
Collapse
Affiliation(s)
- Angelica Escoto
- University of Arizona Department of Molecular and Cellular Biology, Tucson, AZ, 85721, USA
| | - Ryan Hecksel
- University of Arizona Department of Molecular and Cellular Biology, Tucson, AZ, 85721, USA
| | - Chance Parkinson
- University of Arizona Department of Molecular and Cellular Biology, Tucson, AZ, 85721, USA
| | - Sara Crane
- University of Arizona Department of Molecular and Cellular Biology, Tucson, AZ, 85721, USA
| | - Benjamin Atwell
- University of Arizona Department of Molecular and Cellular Biology, Tucson, AZ, 85721, USA
| | - Shyanne King
- University of Arizona Department of Molecular and Cellular Biology, Tucson, AZ, 85721, USA
| | - Daniela Ortiz Chavez
- University of Arizona Department of Molecular and Cellular Biology, Tucson, AZ, 85721, USA
| | - Alison Jannuzi
- University of Arizona Department of Molecular and Cellular Biology, Tucson, AZ, 85721, USA
| | - Barbara Sands
- University of Arizona Department of Molecular and Cellular Biology, Tucson, AZ, 85721, USA
| | - Benjamin G Bitler
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of Colorado School of Medicine, Aurora, CO, 80045, USA
| | - Todd A Fehniger
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Andrew L Paek
- University of Arizona Department of Molecular and Cellular Biology, Tucson, AZ, 85721, USA
- University of Arizona Cancer Center, Tucson, AZ, 85721, USA
| | - Megha Padi
- University of Arizona Department of Molecular and Cellular Biology, Tucson, AZ, 85721, USA.
- University of Arizona Cancer Center, Tucson, AZ, 85721, USA.
| | - Joyce Schroeder
- University of Arizona Department of Molecular and Cellular Biology, Tucson, AZ, 85721, USA.
- University of Arizona Cancer Center, Tucson, AZ, 85721, USA.
- BIO5 Institute, Tucson, AZ, 85721, USA.
| |
Collapse
|
2
|
Dhiman S, Manoj N, Liput M, Sangwan A, Diehl J, Balcerak A, Sudhakar S, Augustyniak J, Jornet JM, Bae Y, Stachowiak EK, Dutta A, Stachowiak MK. Systems Genome: Coordinated Gene Activity Networks, Recurring Coordination Modules, and Genome Homeostasis in Developing Neurons. Int J Mol Sci 2024; 25:5647. [PMID: 38891836 PMCID: PMC11171963 DOI: 10.3390/ijms25115647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Revised: 05/02/2024] [Accepted: 05/10/2024] [Indexed: 06/21/2024] Open
Abstract
As human progenitor cells differentiate into neurons, the activities of many genes change; these changes are maintained within a narrow range, referred to as genome homeostasis. This process, which alters the synchronization of the entire expressed genome, is distorted in neurodevelopmental diseases such as schizophrenia. The coordinated gene activity networks formed by altering sets of genes comprise recurring coordination modules, governed by the entropy-controlling action of nuclear FGFR1, known to be associated with DNA topology. These modules can be modeled as energy-transferring circuits, revealing that genome homeostasis is maintained by reducing oscillations (noise) in gene activity while allowing gene activity changes to be transmitted across networks; this occurs more readily in neuronal committed cells than in neural progenitors. These findings advance a model of an "entangled" global genome acting as a flexible, coordinated homeostatic system that responds to developmental signals, is governed by nuclear FGFR1, and is reprogrammed in disease.
Collapse
Affiliation(s)
- Siddhartha Dhiman
- Department of Biomedical Engineering, University at Buffalo, Buffalo, NY 14228, USA; (S.D.); (A.D.)
| | - Namya Manoj
- Department of Pathology and Anatomical Sciences, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14228, USA; (N.M.); (M.L.); (J.D.); (A.B.); (S.S.); (J.A.); (Y.B.); (E.K.S.)
| | - Michal Liput
- Department of Pathology and Anatomical Sciences, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14228, USA; (N.M.); (M.L.); (J.D.); (A.B.); (S.S.); (J.A.); (Y.B.); (E.K.S.)
- Mossakowski Medical Research Center, Stem Cell Bioengineering Department, Polish Academy of Sciences, Pawinskiego Str., 02-106 Warsaw, Poland
| | - Amit Sangwan
- Department of Electrical Engineering, Northeastern University, Boston, MA 02115, USA; (A.S.); (J.M.J.)
| | - Justin Diehl
- Department of Pathology and Anatomical Sciences, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14228, USA; (N.M.); (M.L.); (J.D.); (A.B.); (S.S.); (J.A.); (Y.B.); (E.K.S.)
| | - Anna Balcerak
- Department of Pathology and Anatomical Sciences, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14228, USA; (N.M.); (M.L.); (J.D.); (A.B.); (S.S.); (J.A.); (Y.B.); (E.K.S.)
| | - Sneha Sudhakar
- Department of Pathology and Anatomical Sciences, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14228, USA; (N.M.); (M.L.); (J.D.); (A.B.); (S.S.); (J.A.); (Y.B.); (E.K.S.)
| | - Justyna Augustyniak
- Department of Pathology and Anatomical Sciences, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14228, USA; (N.M.); (M.L.); (J.D.); (A.B.); (S.S.); (J.A.); (Y.B.); (E.K.S.)
- Mossakowski Medical Research Center, Stem Cell Bioengineering Department, Polish Academy of Sciences, Pawinskiego Str., 02-106 Warsaw, Poland
| | - Josep M. Jornet
- Department of Electrical Engineering, Northeastern University, Boston, MA 02115, USA; (A.S.); (J.M.J.)
| | - Yongho Bae
- Department of Pathology and Anatomical Sciences, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14228, USA; (N.M.); (M.L.); (J.D.); (A.B.); (S.S.); (J.A.); (Y.B.); (E.K.S.)
| | - Ewa K. Stachowiak
- Department of Pathology and Anatomical Sciences, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14228, USA; (N.M.); (M.L.); (J.D.); (A.B.); (S.S.); (J.A.); (Y.B.); (E.K.S.)
| | - Anirban Dutta
- Department of Biomedical Engineering, University at Buffalo, Buffalo, NY 14228, USA; (S.D.); (A.D.)
- Institute of Metabolism and Systems Research, Birmingham Research Park, Birmingham B15 2SQ, UK
| | - Michal K. Stachowiak
- Department of Pathology and Anatomical Sciences, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14228, USA; (N.M.); (M.L.); (J.D.); (A.B.); (S.S.); (J.A.); (Y.B.); (E.K.S.)
| |
Collapse
|
3
|
Wishart TFL, Lovicu FJ. Heparan sulfate proteoglycans (HSPGs) of the ocular lens. Prog Retin Eye Res 2023; 93:101118. [PMID: 36068128 DOI: 10.1016/j.preteyeres.2022.101118] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 08/22/2022] [Accepted: 08/24/2022] [Indexed: 11/17/2022]
Abstract
Heparan sulfate proteoglycans (HSPGs) reside in most cells; on their surface, in the pericellular milieu and/or extracellular matrix. In the eye, HSPGs can orchestrate the activity of key signalling molecules found in the ocular environment that promote its development and homeostasis. To date, our understanding of the specific roles played by individual HSPG family members, and the heterogeneity of their associated sulfated HS chains, is in its infancy. The crystalline lens is a relatively simple and well characterised ocular tissue that provides an ideal stage to showcase and model the expression and unique roles of individual HSPGs. Individual HSPG core proteins are differentially localised to eye tissues in a temporal and spatial developmental- and cell-type specific manner, and their loss or functional disruption results in unique phenotypic outcomes for the lens, and other ocular tissues. More recent work has found that different HS sulfation enzymes are also presented in a cell- and tissue-specific manner, and that disruption of these different sulfation patterns affects specific HS-protein interactions. Not surprisingly, these sulfated HS chains have also been reported to be required for lens and eye development, with dysregulation of HS chain structure and function leading to pathogenesis and eye-related phenotypes. In the lens, HSPGs undergo significant and specific changes in expression and function that can drive pathology, or in some cases, promote tissue repair. As master signalling regulators, HSPGs may one day serve as valuable biomarkers, and even as putative targets for the development of novel therapeutics, not only for the eye but for many other systemic pathologies.
Collapse
Affiliation(s)
- Tayler F L Wishart
- Molecular and Cellular Biomedicine, School of Medical Sciences, The University of Sydney, NSW, Australia.
| | - Frank J Lovicu
- Molecular and Cellular Biomedicine, School of Medical Sciences, The University of Sydney, NSW, Australia; Save Sight Institute, The University of Sydney, NSW, Australia.
| |
Collapse
|
4
|
Li G, Choi JE, Kryczek I, Sun Y, Liao P, Li S, Wei S, Grove S, Vatan L, Nelson R, Schaefer G, Allen SG, Sankar K, Fecher LA, Mendiratta-Lala M, Frankel TL, Qin A, Waninger JJ, Tezel A, Alva A, Lao CD, Ramnath N, Cieslik M, Harms PW, Green MD, Chinnaiyan AM, Zou W. Intersection of immune and oncometabolic pathways drives cancer hyperprogression during immunotherapy. Cancer Cell 2023; 41:304-322.e7. [PMID: 36638784 PMCID: PMC10286807 DOI: 10.1016/j.ccell.2022.12.008] [Citation(s) in RCA: 50] [Impact Index Per Article: 50.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 11/07/2022] [Accepted: 12/20/2022] [Indexed: 01/13/2023]
Abstract
Immune checkpoint blockade (ICB) can produce durable responses against cancer. We and others have found that a subset of patients experiences paradoxical rapid cancer progression during immunotherapy. It is poorly understood how tumors can accelerate their progression during ICB. In some preclinical models, ICB causes hyperprogressive disease (HPD). While immune exclusion drives resistance to ICB, counterintuitively, patients with HPD and complete response (CR) following ICB manifest comparable levels of tumor-infiltrating CD8+ T cells and interferon γ (IFNγ) gene signature. Interestingly, patients with HPD but not CR exhibit elevated tumoral fibroblast growth factor 2 (FGF2) and β-catenin signaling. In animal models, T cell-derived IFNγ promotes tumor FGF2 signaling, thereby suppressing PKM2 activity and decreasing NAD+, resulting in reduction of SIRT1-mediated β-catenin deacetylation and enhanced β-catenin acetylation, consequently reprograming tumor stemness. Targeting the IFNγ-PKM2-β-catenin axis prevents HPD in preclinical models. Thus, the crosstalk of core immunogenic, metabolic, and oncogenic pathways via the IFNγ-PKM2-β-catenin cascade underlies ICB-associated HPD.
Collapse
Affiliation(s)
- Gaopeng Li
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA; Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan Rogel Cancer Center, Ann Arbor, MI, USA
| | - Jae Eun Choi
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA; Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Ilona Kryczek
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA; Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan Rogel Cancer Center, Ann Arbor, MI, USA
| | - Yilun Sun
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA; Department of Biostatistics, University of Michigan, Ann Arbor, MI, USA
| | - Peng Liao
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA; Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan Rogel Cancer Center, Ann Arbor, MI, USA
| | - Shasha Li
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA; Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan Rogel Cancer Center, Ann Arbor, MI, USA
| | - Shuang Wei
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA; Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan Rogel Cancer Center, Ann Arbor, MI, USA
| | - Sara Grove
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA; Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan Rogel Cancer Center, Ann Arbor, MI, USA
| | - Linda Vatan
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA; Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan Rogel Cancer Center, Ann Arbor, MI, USA
| | - Reagan Nelson
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA
| | - Grace Schaefer
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA
| | - Steven G Allen
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA
| | - Kamya Sankar
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Leslie A Fecher
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | | | | | - Angel Qin
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Jessica J Waninger
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA; Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA
| | - Alangoya Tezel
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA
| | - Ajjai Alva
- Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan Rogel Cancer Center, Ann Arbor, MI, USA; Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Christopher D Lao
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Nithya Ramnath
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA; Veterans Affairs Ann Arbor Healthcare System, Ann Arbor, MI, USA
| | - Marcin Cieslik
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA; Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA; Department of Computational Medicine & Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Paul W Harms
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Michael D Green
- Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan Rogel Cancer Center, Ann Arbor, MI, USA; Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA; Veterans Affairs Ann Arbor Healthcare System, Ann Arbor, MI, USA; Graduate Program in Immunology, University of Michigan, Ann Arbor, MI, USA; Graduate Program in Cancer Biology, University of Michigan, Ann Arbor, MI, USA.
| | - Arul M Chinnaiyan
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA; Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA; Howard Hughes Medical Institute, University of Michigan, Ann Arbor, MI, USA
| | - Weiping Zou
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA; Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan Rogel Cancer Center, Ann Arbor, MI, USA; Department of Pathology, University of Michigan, Ann Arbor, MI, USA; Graduate Program in Immunology, University of Michigan, Ann Arbor, MI, USA; Graduate Program in Cancer Biology, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
5
|
Kim JA, Berlow NE, Lathara M, Bharathy N, Martin LR, Purohit R, Cleary MM, Liu Q, Michalek JE, Srinivasa G, Cole BL, Chen SD, Keller C. Sensitization of osteosarcoma to irradiation by targeting nuclear FGFR1. Biochem Biophys Res Commun 2022; 621:101-108. [DOI: 10.1016/j.bbrc.2022.07.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 06/22/2022] [Accepted: 07/01/2022] [Indexed: 11/30/2022]
|
6
|
Suh J, Kim DH, Kim SJ, Cho NC, Lee YH, Jang JH, Surh YJ. Nuclear Localization of Fibroblast Growth Factor Receptor 1 in Breast Cancer Cells Interacting with Cancer Associated Fibroblasts. J Cancer Prev 2022; 27:68-76. [PMID: 35419302 PMCID: PMC8984647 DOI: 10.15430/jcp.2022.27.1.68] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 02/22/2022] [Accepted: 02/25/2022] [Indexed: 12/03/2022] Open
Abstract
Cancer-associated fibroblasts (CAFs) represent a major component of the tumor microenvironment and interplay with cancer cells by secreting cytokines, growth factors and extracellular matrix proteins. When estrogen receptor-negative breast cancer MDA-MB-231 cells were treated with the CAF-conditioned medium (CAF-CM), Akt and STAT3 involved in cell proliferation and survival were activated through phosphorylation. CAFs secrete fibroblast growth factor 2 (FGF2), thereby stimulating breast cancer cell progression. Akt activation induced by CAF-CM in MDA-MB-231 cells was abolished when FGF2-neutralizing antibody was added. Treatment of MDA-MB-231 cells directly with FGF2 enhanced the phosphorylation of Akt and the FGF receptor (FGFR) substrate, FRS2α. These events were abrogated by siRNA-mediated silencing of FGFR1. In a xenograft mouse model, co-injection of MDA-MB-231 cells with activated fibroblasts expressing FGF2 dramatically enhanced activation of Akt. Stable knockdown of FGFR1 blunted Akt phosphorylation in xenograft tumors. MDA-MB-231 cells co-cultured with CAFs or directly stimulated with FGF2 exhibited enhanced nuclear localization of FGFR1. Notably, FGF2 stimulation produced reactive oxygen species (ROS) accumulation in MDA-MB-231 cells, and FGF2-induced nuclear accumulation of FGFR1 was abrogated by the ROS scavenging agent, N-acetylcysteine.
Collapse
Affiliation(s)
- Jinyoung Suh
- Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Korea
| | - Do-Hee Kim
- Department of Chemistry, College of Convergence and Integrated Science, Kyonggi University, Suwon, Korea
| | - Su-Jung Kim
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, Korea
| | - Nam-Chul Cho
- Drug Information Platfom Center, Korea Chemical Bank, Korea Research Institute of Chemical Technology, Daejeon, Korea
| | - Yeon-Hwa Lee
- Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Korea
| | - Jeong-Hoon Jang
- Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Korea
| | - Young-Joon Surh
- Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Korea
- Cancer Research Institute, Seoul National University, Seoul, Korea
| |
Collapse
|
7
|
Regulation of FGF-2, FGF-18 and Transcription Factor Activity by Perlecan in the Maturational Development of Transitional Rudiment and Growth Plate Cartilages and in the Maintenance of Permanent Cartilage Homeostasis. Int J Mol Sci 2022; 23:ijms23041934. [PMID: 35216048 PMCID: PMC8872392 DOI: 10.3390/ijms23041934] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 01/24/2022] [Accepted: 02/01/2022] [Indexed: 12/11/2022] Open
Abstract
The aim of this study was to highlight the roles of perlecan in the regulation of the development of the rudiment developmental cartilages and growth plate cartilages, and also to show how perlecan maintains permanent articular cartilage homeostasis. Cartilage rudiments are transient developmental templates containing chondroprogenitor cells that undergo proliferation, matrix deposition, and hypertrophic differentiation. Growth plate cartilage also undergoes similar changes leading to endochondral bone formation, whereas permanent cartilage is maintained as an articular structure and does not undergo maturational changes. Pericellular and extracellular perlecan-HS chains interact with growth factors, morphogens, structural matrix glycoproteins, proteases, and inhibitors to promote matrix stabilization and cellular proliferation, ECM remodelling, and tissue expansion. Perlecan has mechanotransductive roles in cartilage that modulate chondrocyte responses in weight-bearing environments. Nuclear perlecan may modulate chromatin structure and transcription factor access to DNA and gene regulation. Snail-1, a mesenchymal marker and transcription factor, signals through FGFR-3 to promote chondrogenesis and maintain Acan and type II collagen levels in articular cartilage, but prevents further tissue expansion. Pre-hypertrophic growth plate chondrocytes also express high Snail-1 levels, leading to cessation of Acan and CoI2A1 synthesis and appearance of type X collagen. Perlecan differentially regulates FGF-2 and FGF-18 to maintain articular cartilage homeostasis, rudiment and growth plate cartilage growth, and maturational changes including mineralization, contributing to skeletal growth.
Collapse
|
8
|
Mahmood HTNA, Tomas Bort E, Walker AJ, Grose RP, Chioni AM. FGF signalling facilitates cervical cancer progression. FEBS J 2021; 289:3440-3456. [PMID: 34951738 DOI: 10.1111/febs.16331] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 11/26/2021] [Accepted: 12/21/2021] [Indexed: 12/18/2022]
Abstract
Cervical cancer is one of the most frequently diagnosed cancers in women worldwide. While cervical cancer is caused by human papillomavirus (HPV), not all females infected with HPV develop the disease, suggesting that other factors might facilitate its progression. Growing evidence supports the involvement of the fibroblast growth factor receptor (FGFR) axis in several cancers, including gynecological. However, for cervical cancer, the molecular mechanisms that underpin the disease remain poorly understood, including the role of FGFR signaling. The aim of this study was to investigate FGF(R) signaling in cervical cancer through bioinformatic analysis of cell line and patient data and through detailed expression profiling, manipulation of the FGFR axis, and downstream phenotypic analysis in cell lines (HeLa, SiHa, and CaSki). Expression (protein and mRNA) analysis demonstrated that FGFR1b/c, FGFR2b/c, FGFR4, FGF2, FGF4, and FGF7 were expressed in all three lines. Interestingly, FGFR1 and 2 localized to the nucleus, supporting that nuclear FGFRs could act as transcription factors. Importantly, 2D and 3D cell cultures demonstrated that FGFR activation can facilitate cell functions correlated with invasive disease. Collectively, this study supports an association between FGFR signaling and cervical cancer progression, laying the foundations for the development of therapeutic approaches targeting FGFR in this disease.
Collapse
Affiliation(s)
| | - Elena Tomas Bort
- Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, UK
| | - Anthony J Walker
- School of Life Sciences Pharmacy and Chemistry, Kingston University, Kingston upon Thames, UK
| | - Richard P Grose
- Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, UK
| | - Athina-Myrto Chioni
- School of Life Sciences Pharmacy and Chemistry, Kingston University, Kingston upon Thames, UK
| |
Collapse
|
9
|
Ogawa Y, Imamoto N. Methods to separate nuclear soluble fractions reflecting localizations in living cells. iScience 2021; 24:103503. [PMID: 34934922 DOI: 10.1016/j.isci.2021.103503] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 08/15/2021] [Accepted: 11/22/2021] [Indexed: 12/16/2022] Open
Abstract
To understand various intranuclear functions, it is important to know when, what, and how proteins enter the nucleus. Although many methods and commercial kits for nuclear fractionation have been developed, there are still no methods for obtaining a complete nuclear proteome. Soluble nuclear proteins are often lost during fractionation. We developed remarkably improved methods to obtain nuclear soluble fractions by optimizing the conditions of selective permeabilization of the plasma membrane. As a result, 10 million cells could be separated into the cytoplasmic and nuclear soluble fractions more precisely in a 1.5-mL test tube. Moreover, the addition of an inhibitor to prevent leakage from the nucleus retained small proteins in the nucleus. Because of the simple protocols and easy application for multiple samples, our methods are expected to be applied to various studies on spatiotemporal changes of dynamic nuclear proteins, such as signal transduction.
Collapse
Affiliation(s)
- Yutaka Ogawa
- Cellular Dynamics Laboratory, RIKEN Cluster for Pioneering Research, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | - Naoko Imamoto
- Cellular Dynamics Laboratory, RIKEN Cluster for Pioneering Research, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| |
Collapse
|
10
|
Wishart TFL, Lovicu FJ. An Atlas of Heparan Sulfate Proteoglycans in the Postnatal Rat Lens. Invest Ophthalmol Vis Sci 2021; 62:5. [PMID: 34730792 PMCID: PMC8572486 DOI: 10.1167/iovs.62.14.5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Purpose The arrangement of lens cells is regulated by ocular growth factors. Although the effects of these inductive molecules on lens cell behavior (proliferation, survival, and fiber differentiation) are well-characterized, the precise mechanisms underlying the regulation of growth factor-mediated signaling in lens remains elusive. Increasing evidence highlights the importance of heparan sulfate proteoglycans (HSPGs) for the signaling regulation of growth factors; however, the identity of the different lens HSPGs and the specific roles they play in lens biology are still unknown. Methods Semiquantitative real-time (RT)‐PCR and immunolabeling were used to characterize the spatial distribution of all known HSPG core proteins and their associated glycosaminoglycans (heparan and chondroitin sulfate) in the postnatal rat lens. Fibroblast growth factor (FGF)-2-treated lens epithelial explants, cultured in the presence of Surfen (an inhibitor of heparan sulfate [HS]-growth factor binding interactions) were used to investigate the requirement for HS in FGF-2-induced proliferation, fiber differentiation, and ERK1/2-signaling. Results The lens expresses all HSPGs. These HSPGs are differentially localized to distinct functional regions of the lens. In vitro, inhibition of HS-sulfation with Surfen blocked FGF-2-mediated ERK1/2-signaling associated with lens epithelial cell proliferation and fiber differentiation, highlighting that these cellular processes are dependent on HS. Conclusions These findings support a requirement for HSPGs in FGF-2 driven lens cell proliferation and fiber differentiation. The identification of specific HSPG core proteins in key functional lens regions, and the divergent expression patterns of closely related HSPGs, suggests that different HSPGs may differentially regulate growth factor signaling networks leading to specific biological events involved in lens growth and maintenance.
Collapse
Affiliation(s)
- Tayler F L Wishart
- School of Medical Sciences, The University of Sydney, New South Wales, Australia
| | - Frank J Lovicu
- School of Medical Sciences, The University of Sydney, New South Wales, Australia.,Save Sight Institute, The University of Sydney, New South Wales, Australia
| |
Collapse
|
11
|
Klimaschewski L, Claus P. Fibroblast Growth Factor Signalling in the Diseased Nervous System. Mol Neurobiol 2021; 58:3884-3902. [PMID: 33860438 PMCID: PMC8280051 DOI: 10.1007/s12035-021-02367-0] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 03/19/2021] [Indexed: 12/12/2022]
Abstract
Fibroblast growth factors (FGFs) act as key signalling molecules in brain development, maintenance, and repair. They influence the intricate relationship between myelinating cells and axons as well as the association of astrocytic and microglial processes with neuronal perikarya and synapses. Advances in molecular genetics and imaging techniques have allowed novel insights into FGF signalling in recent years. Conditional mouse mutants have revealed the functional significance of neuronal and glial FGF receptors, not only in tissue protection, axon regeneration, and glial proliferation but also in instant behavioural changes. This review provides a summary of recent findings regarding the role of FGFs and their receptors in the nervous system and in the pathogenesis of major neurological and psychiatric disorders.
Collapse
Affiliation(s)
- Lars Klimaschewski
- Department of Anatomy, Histology and Embryology, Institute of Neuroanatomy, Medical University of Innsbruck, Innsbruck, Austria.
| | - Peter Claus
- Institute of Neuroanatomy and Cell Biology, Hannover Medical School, Hannover, Germany
- Center for Systems Neuroscience, Hannover, Germany
| |
Collapse
|
12
|
What Are the Potential Roles of Nuclear Perlecan and Other Heparan Sulphate Proteoglycans in the Normal and Malignant Phenotype. Int J Mol Sci 2021; 22:ijms22094415. [PMID: 33922532 PMCID: PMC8122901 DOI: 10.3390/ijms22094415] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 04/19/2021] [Accepted: 04/19/2021] [Indexed: 12/27/2022] Open
Abstract
The recent discovery of nuclear and perinuclear perlecan in annulus fibrosus and nucleus pulposus cells and its known matrix stabilizing properties in tissues introduces the possibility that perlecan may also have intracellular stabilizing or regulatory roles through interactions with nuclear envelope or cytoskeletal proteins or roles in nucleosomal-chromatin organization that may regulate transcriptional factors and modulate gene expression. The nucleus is a mechano-sensor organelle, and sophisticated dynamic mechanoresponsive cytoskeletal and nuclear envelope components support and protect the nucleus, allowing it to perceive and respond to mechano-stimulation. This review speculates on the potential roles of perlecan in the nucleus based on what is already known about nuclear heparan sulphate proteoglycans. Perlecan is frequently found in the nuclei of tumour cells; however, its specific role in these diseased tissues is largely unknown. The aim of this review is to highlight probable roles for this intriguing interactive regulatory proteoglycan in the nucleus of normal and malignant cell types.
Collapse
|
13
|
Döring P, Calvisi DF, Dombrowski F. Nuclear ErbB2 expression in hepatocytes in liver disease. Virchows Arch 2020; 478:309-318. [PMID: 32591879 PMCID: PMC7969555 DOI: 10.1007/s00428-020-02871-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 06/02/2020] [Accepted: 06/12/2020] [Indexed: 12/03/2022]
Abstract
ErbB2 is a prominent representative of the epidermal growth factor receptors that mainly attract attention as oncogenic drivers and therapeutic targets in cancer. Besides transmembrane signaling, ErbB2 may also translocate into the nucleus and mediate distinct nuclear signaling effects including DNA repair and cell cycle arrest. Unexpectedly, we found nuclear ErbB2 expression in human hepatocytes in various liver diseases so we aimed to investigate the characteristics of liver disease leading to nuclear ErbB2 translocation. The immunohistochemical pattern of ErbB2 staining was analyzed in 1125 liver biopsy samples from patients with hepatic dysfunction. Further signaling and metabolic markers were analyzed by immunohistochemistry in selected liver biopsy samples. We found a cytoplasmic and nuclear ErbB2 expression in hepatocytes from different disease conditions with the strongest expression detected in alcoholic steatohepatitis. Nuclear ErbB2 positivity significantly correlated with histologic parameters of hepatocellular damage including inflammatory activity in steatohepatitis, hepatocellular ballooning, and cholestasis. ErbB2 overexpressing hepatocytes revealed an increase of phospho-STAT3, a downstream effector of nuclear ErbB2 signaling. Notably, we observed in nuclear ErbB2-positive hepatocytes a downregulation of estrogen receptor expression. In alcoholic steatohepatitis and other toxic liver diseases, hepatocytes revealed a nuclear ErbB2 expression implying a so far unknown mechanism in hepatocytes upon cellular stress that might lead to resistance to cell death. Nuclear ErbB2-positive hepatocytes showed downregulation of estrogen receptor expression and increased levels of pSTAT3, which are signs of functionality of nuclear ErbB2 signaling. Furthermore, analysis of hepatocellular ErbB2 expression could serve as helpful tool for diagnosis of liver disease.
Collapse
Affiliation(s)
- Paula Döring
- Institute of Pathology, Universitätsmedizin Greifswald, Friedrich-Loeffler-Straße 23e, 17475, Greifswald, Germany.
| | - Diego F Calvisi
- Institute of Pathology, University of Regensburg, Regensburg, Germany
| | - Frank Dombrowski
- Institute of Pathology, Universitätsmedizin Greifswald, Friedrich-Loeffler-Straße 23e, 17475, Greifswald, Germany
| |
Collapse
|
14
|
Figueroa V, Rodríguez MS, Lanari C, Lamb CA. Nuclear action of FGF members in endocrine-related tissues and cancer: Interplay with steroid receptor pathways. Steroids 2019; 152:108492. [PMID: 31513818 DOI: 10.1016/j.steroids.2019.108492] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Revised: 08/27/2019] [Accepted: 09/05/2019] [Indexed: 01/09/2023]
Abstract
Dysregulation of the fibroblast growth factors/fibroblast growth factor receptor (FGF/FGFR) pathway has been implicated in a wide range of human disorders and several members have been localized in the nuclear compartment. Hormone-activated steroid receptors or ligand independent activated receptors form nuclear complexes that activate gene transcription. This review aims to highlight the interplay between the steroid receptor and the FGF/FGFR pathways and focuses on the current knowledge on nuclear action of FGF members in endocrine-related tissues and cancer. The nuclear trafficking and targets of FGF/FGFR members and the available evidence on the interplay with steroid hormones and receptors is described. Finally, the data on aberrant FGF/FGFR signaling is summarized and the nuclear action of FGF members on endocrine resistant breast cancer is highlighted. Identifying the mechanisms underlying FGF-induced endocrine resistance will be important to understand how to efficiently target endocrine-related diseases and even enhance or restore endocrine sensitivity in hormone receptor positive tumors.
Collapse
Affiliation(s)
- Virginia Figueroa
- Instituto de Biología y Medicina Experimental (IBYME-CONICET), Vuelta de Obligado 2490, Buenos Aires 1428, Argentina
| | - María Sol Rodríguez
- Instituto de Biología y Medicina Experimental (IBYME-CONICET), Vuelta de Obligado 2490, Buenos Aires 1428, Argentina
| | - Claudia Lanari
- Instituto de Biología y Medicina Experimental (IBYME-CONICET), Vuelta de Obligado 2490, Buenos Aires 1428, Argentina
| | - Caroline Ana Lamb
- Instituto de Biología y Medicina Experimental (IBYME-CONICET), Vuelta de Obligado 2490, Buenos Aires 1428, Argentina.
| |
Collapse
|
15
|
Chiusa M, Hu W, Liao HJ, Su Y, Borza CM, de Caestecker MP, Skrypnyk NI, Fogo AB, Pedchenko V, Li X, Zhang MZ, Hudson BG, Basak T, Vanacore RM, Zent R, Pozzi A. The Extracellular Matrix Receptor Discoidin Domain Receptor 1 Regulates Collagen Transcription by Translocating to the Nucleus. J Am Soc Nephrol 2019; 30:1605-1624. [PMID: 31383731 PMCID: PMC6727269 DOI: 10.1681/asn.2018111160] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2018] [Accepted: 05/20/2019] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND The discoidin domain receptor 1 (DDR1) is activated by collagens, upregulated in injured and fibrotic kidneys, and contributes to fibrosis by regulating extracellular matrix production, but how DDR1 controls fibrosis is poorly understood. DDR1 is a receptor tyrosine kinase (RTK). RTKs can translocate to the nucleus via a nuclear localization sequence (NLS) present on the receptor itself or a ligand it is bound to. In the nucleus, RTKs regulate gene expression by binding chromatin directly or by interacting with transcription factors. METHODS To determine whether DDR1 translocates to the nucleus and whether this event is mediated by collagen-induced DDR1 activation, we generated renal cells expressing wild-type or mutant forms of DDR1 no longer able to bind collagen. Then, we determined the location of the DDR1 upon collagen stimulation. Using both biochemical assays and immunofluorescence, we analyzed the steps involved in DDR1 nuclear translocation. RESULTS We show that although DDR1 and its natural ligand, collagen, lack an NLS, DDR1 is present in the nucleus of injured human and mouse kidney proximal tubules. We show that DDR1 nuclear translocation requires collagen-mediated receptor activation and interaction of DDR1 with SEC61B, a component of the Sec61 translocon, and nonmuscle myosin IIA and β-actin. Once in the nucleus, DDR1 binds to chromatin to increase the transcription of collagen IV, a major collagen upregulated in fibrosis. CONCLUSIONS These findings reveal a novel mechanism whereby activated DDR1 translates to the nucleus to regulate synthesis of profibrotic molecules.
Collapse
Affiliation(s)
- Manuel Chiusa
- Division of Nephrology and Hypertension, Department of Medicine, and
| | - Wen Hu
- Division of Nephrology and Hypertension, Department of Medicine, and
| | - Hong-Jun Liao
- Division of Nephrology and Hypertension, Department of Medicine, and
| | - Yan Su
- Division of Nephrology and Hypertension, Department of Medicine, and
| | - Corina M Borza
- Division of Nephrology and Hypertension, Department of Medicine, and
| | | | | | - Agnes B Fogo
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee; and
| | - Vadim Pedchenko
- Division of Nephrology and Hypertension, Department of Medicine, and
| | - Xiyue Li
- Division of Nephrology and Hypertension, Department of Medicine, and
| | - Ming-Zhi Zhang
- Division of Nephrology and Hypertension, Department of Medicine, and
| | - Billy G Hudson
- Division of Nephrology and Hypertension, Department of Medicine, and
| | - Trayambak Basak
- Division of Nephrology and Hypertension, Department of Medicine, and
| | | | - Roy Zent
- Division of Nephrology and Hypertension, Department of Medicine, and
- Department of Veterans Affairs, Nashville, Tennessee
| | - Ambra Pozzi
- Division of Nephrology and Hypertension, Department of Medicine, and
- Department of Veterans Affairs, Nashville, Tennessee
| |
Collapse
|
16
|
The NAE Pathway: Autobahn to the Nucleus for Cell Surface Receptors. Cells 2019; 8:cells8080915. [PMID: 31426451 PMCID: PMC6721735 DOI: 10.3390/cells8080915] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 08/13/2019] [Accepted: 08/13/2019] [Indexed: 12/19/2022] Open
Abstract
Various growth factors and full-length cell surface receptors such as EGFR are translocated from the cell surface to the nucleoplasm, baffling cell biologists to the mechanisms and functions of this process. Elevated levels of nuclear EGFR correlate with poor prognosis in various cancers. In recent years, nuclear EGFR has been implicated in regulating gene transcription, cell proliferation and DNA damage repair. Different models have been proposed to explain how the receptors are transported into the nucleus. However, a clear consensus has yet to be reached. Recently, we described the nuclear envelope associated endosomes (NAE) pathway, which delivers EGFR from the cell surface to the nucleus. This pathway involves transport, docking and fusion of NAEs with the outer membrane of the nuclear envelope. EGFR is then presumed to be transported through the nuclear pore complex, extracted from membranes and solubilised. The SUN1/2 nuclear envelope proteins, Importin-beta, nuclear pore complex proteins and the Sec61 translocon have been implicated in the process. While this framework can explain the cell surface to nucleus traffic of EGFR and other cell surface receptors, it raises several questions that we consider in this review, together with implications for health and disease.
Collapse
|
17
|
Tuzon CT, Rigueur D, Merrill AE. Nuclear Fibroblast Growth Factor Receptor Signaling in Skeletal Development and Disease. Curr Osteoporos Rep 2019; 17:138-146. [PMID: 30982184 PMCID: PMC8221190 DOI: 10.1007/s11914-019-00512-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
PURPOSE OF REVIEW Fibroblast growth factor receptor (FGFR) signaling regulates proliferation and differentiation during development and homeostasis. While membrane-bound FGFRs play a central role in these processes, the function of nuclear FGFRs is also critical. Here, we highlight mechanisms for nuclear FGFR translocation and the effects of nuclear FGFRs on skeletal development and disease. RECENT FINDINGS Full-length FGFRs, internalized by endocytosis, enter the nucleus through β-importin-dependent mechanisms that recognize the nuclear localization signal within FGFs. Alternatively, soluble FGFR intracellular fragments undergo nuclear translocation following their proteolytic release from the membrane. FGFRs enter the nucleus during the cellular transition between proliferation and differentiation. Once nuclear, FGFRs interact with chromatin remodelers to alter the epigenetic state and transcription of their target genes. Dysregulation of nuclear FGFR is linked to the etiology of congenital skeletal disorders and neoplastic transformation. Revealing the activities of nuclear FGFR will advance our understanding of 20 congenital skeletal disorders caused by FGFR mutations, as well as FGFR-related cancers.
Collapse
Affiliation(s)
- Creighton T Tuzon
- Center for Craniofacial Molecular Biology, Herman Ostrow School of Dentistry, University of Southern California, Los Angeles, CA, 90033, USA
| | - Diana Rigueur
- Center for Craniofacial Molecular Biology, Herman Ostrow School of Dentistry, University of Southern California, Los Angeles, CA, 90033, USA
| | - Amy E Merrill
- Center for Craniofacial Molecular Biology, Herman Ostrow School of Dentistry, University of Southern California, Los Angeles, CA, 90033, USA.
- Department of Biochemistry and Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA.
| |
Collapse
|
18
|
di Martino E, Alder O, Hurst CD, Knowles MA. ETV5 links the FGFR3 and Hippo signalling pathways in bladder cancer. Sci Rep 2019; 9:5740. [PMID: 30952872 PMCID: PMC6450944 DOI: 10.1038/s41598-018-36456-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Accepted: 11/14/2018] [Indexed: 12/29/2022] Open
Abstract
Activating mutations of fibroblast growth factor receptor 3 (FGFR3) are common in urothelial carcinoma of the bladder (UC). Silencing or inhibition of mutant FGFR3 in bladder cancer cell lines is associated with decreased malignant potential, confirming its important driver role in UC. However, understanding of how FGFR3 activation drives urothelial malignant transformation remains limited. We have previously shown that mutant FGFR3 alters the cell-cell and cell-matrix adhesion properties of urothelial cells, resulting in loss of contact-inhibition of proliferation. In this study, we investigate a transcription factor of the ETS-family, ETV5, as a putative effector of FGFR3 signalling in bladder cancer. We show that FGFR3 signalling induces a MAPK/ERK-mediated increase in ETV5 levels, and that this results in increased level of TAZ, a co-transcriptional regulator downstream of the Hippo signalling pathway involved in cell-contact inhibition. We also demonstrate that ETV5 is a key downstream mediator of the oncogenic effects of mutant FGFR3, as its knockdown in FGFR3-mutant bladder cancer cell lines is associated with reduced proliferation and anchorage-independent growth. Overall this study advances our understanding of the molecular alterations occurring during urothelial malignant transformation and indicates TAZ as a possible therapeutic target in FGFR3-dependent bladder tumours.
Collapse
Affiliation(s)
- Erica di Martino
- University of Leeds, Leeds Institute of Medical Research at St James's, St. James's University Hospital, Leeds, LS9 7TF, UK
| | - Olivia Alder
- University of Leeds, Leeds Institute of Medical Research at St James's, St. James's University Hospital, Leeds, LS9 7TF, UK
| | - Carolyn D Hurst
- University of Leeds, Leeds Institute of Medical Research at St James's, St. James's University Hospital, Leeds, LS9 7TF, UK
| | - Margaret A Knowles
- University of Leeds, Leeds Institute of Medical Research at St James's, St. James's University Hospital, Leeds, LS9 7TF, UK.
| |
Collapse
|
19
|
Membrane-Associated, Not Cytoplasmic or Nuclear, FGFR1 Induces Neuronal Differentiation. Cells 2019; 8:cells8030243. [PMID: 30875802 PMCID: PMC6468866 DOI: 10.3390/cells8030243] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 03/04/2019] [Accepted: 03/08/2019] [Indexed: 01/22/2023] Open
Abstract
The intracellular transport of receptor tyrosine kinases results in the differential activation of various signaling pathways. In this study, optogenetic stimulation of fibroblast growth factor receptor type 1 (FGFR1) was performed to study the effects of subcellular targeting of receptor kinases on signaling and neurite outgrowth. The catalytic domain of FGFR1 fused to the algal light-oxygen-voltage-sensing (LOV) domain was directed to different cellular compartments (plasma membrane, cytoplasm and nucleus) in human embryonic kidney (HEK293) and pheochromocytoma (PC12) cells. Blue light stimulation elevated the pERK and pPLCγ1 levels in membrane-opto-FGFR1-transfected cells similarly to ligand-induced receptor activation; however, no changes in pAKT levels were observed. PC12 cells transfected with membrane-opto-FGFR1 exhibited significantly longer neurites after light stimulation than after growth factor treatment, and significantly more neurites extended from their cell bodies. The activation of cytoplasmic FGFR1 kinase enhanced ERK signaling in HEK293 cells but not in PC12 cells and did not induce neuronal differentiation. The stimulation of FGFR1 kinase in the nucleus also did not result in signaling changes or neurite outgrowth. We conclude that FGFR1 kinase needs to be associated with membranes to induce the differentiation of PC12 cells mainly via ERK activation.
Collapse
|
20
|
Porębska N, Latko M, Kucińska M, Zakrzewska M, Otlewski J, Opaliński Ł. Targeting Cellular Trafficking of Fibroblast Growth Factor Receptors as a Strategy for Selective Cancer Treatment. J Clin Med 2018; 8:jcm8010007. [PMID: 30577533 PMCID: PMC6352210 DOI: 10.3390/jcm8010007] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 12/17/2018] [Accepted: 12/17/2018] [Indexed: 12/13/2022] Open
Abstract
Fibroblast growth factor receptors (FGFRs) in response to fibroblast growth factors (FGFs) transmit signals across the cell membrane, regulating important cellular processes, like differentiation, division, motility, and death. The aberrant activity of FGFRs is often observed in various diseases, especially in cancer. The uncontrolled FGFRs' function may result from their overproduction, activating mutations, or generation of FGFRs' fusion proteins. Besides their typical subcellular localization on the cell surface, FGFRs are often found inside the cells, in the nucleus and mitochondria. The intracellular pool of FGFRs utilizes different mechanisms to facilitate cancer cell survival and expansion. In this review, we summarize the current stage of knowledge about the role of FGFRs in oncogenic processes. We focused on the mechanisms of FGFRs' cellular trafficking-internalization, nuclear translocation, and mitochondrial targeting, as well as their role in carcinogenesis. The subcellular sorting of FGFRs constitutes an attractive target for anti-cancer therapies. The blocking of FGFRs' nuclear and mitochondrial translocation can lead to the inhibition of cancer invasion. Moreover, the endocytosis of FGFRs can serve as a tool for the efficient and highly selective delivery of drugs into cancer cells overproducing these receptors. Here, we provide up to date examples how the cellular sorting of FGFRs can be hijacked for selective cancer treatment.
Collapse
Affiliation(s)
- Natalia Porębska
- Department of Protein Engineering, Faculty of Biotechnology, University of Wrocław, Joliot-Curie 14a, 50-383 Wroclaw, Poland.
| | - Marta Latko
- Department of Protein Engineering, Faculty of Biotechnology, University of Wrocław, Joliot-Curie 14a, 50-383 Wroclaw, Poland.
| | - Marika Kucińska
- Department of Protein Engineering, Faculty of Biotechnology, University of Wrocław, Joliot-Curie 14a, 50-383 Wroclaw, Poland.
| | - Małgorzata Zakrzewska
- Department of Protein Engineering, Faculty of Biotechnology, University of Wrocław, Joliot-Curie 14a, 50-383 Wroclaw, Poland.
| | - Jacek Otlewski
- Department of Protein Engineering, Faculty of Biotechnology, University of Wrocław, Joliot-Curie 14a, 50-383 Wroclaw, Poland.
| | - Łukasz Opaliński
- Department of Protein Engineering, Faculty of Biotechnology, University of Wrocław, Joliot-Curie 14a, 50-383 Wroclaw, Poland.
| |
Collapse
|
21
|
Weddell JC, Imoukhuede PI. Integrative meta-modeling identifies endocytic vesicles, late endosome and the nucleus as the cellular compartments primarily directing RTK signaling. Integr Biol (Camb) 2018; 9:464-484. [PMID: 28436498 DOI: 10.1039/c7ib00011a] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Recently, intracellular receptor signaling has been identified as a key component mediating cell responses for various receptor tyrosine kinases (RTKs). However, the extent each endocytic compartment (endocytic vesicle, early endosome, recycling endosome, late endosome, lysosome and nucleus) contributes to receptor signaling has not been quantified. Furthermore, our understanding of endocytosis and receptor signaling is complicated by cell- or receptor-specific endocytosis mechanisms. Therefore, towards understanding the differential endocytic compartment signaling roles, and identifying how to achieve signal transduction control for RTKs, we delineate how endocytosis regulates RTK signaling. We achieve this via a meta-analysis across eight RTKs, integrating computational modeling with experimentally derived cell (compartment volume, trafficking kinetics and pH) and ligand-receptor (ligand/receptor concentration and interaction kinetics) physiology. Our simulations predict the abundance of signaling from eight RTKs, identifying the following hierarchy in RTK signaling: PDGFRβ > IGFR1 > EGFR > PDGFRα > VEGFR1 > VEGFR2 > Tie2 > FGFR1. We find that endocytic vesicles are the primary cell signaling compartment; over 43% of total receptor signaling occurs within the endocytic vesicle compartment for these eight RTKs. Mechanistically, we found that high RTK signaling within endocytic vesicles may be attributed to their low volume (5.3 × 10-19 L) which facilitates an enriched ligand concentration (3.2 μM per ligand molecule within the endocytic vesicle). Under the analyzed physiological conditions, we identified extracellular ligand concentration as the most sensitive parameter to change; hence the most significant one to modify when regulating absolute compartment signaling. We also found that the late endosome and nucleus compartments are important contributors to receptor signaling, where 26% and 18%, respectively, of average receptor signaling occurs across the eight RTKs. Conversely, we found very low membrane-based receptor signaling, exhibiting <1% of the total receptor signaling for these eight RTKs. Moreover, we found that nuclear translocation, mechanistically, requires late endosomal transport; when we blocked receptor trafficking from late endosomes to the nucleus we found a 57% reduction in nuclear translocation. In summary, our research has elucidated the significance of endocytic vesicles, late endosomes and the nucleus in RTK signal propagation.
Collapse
Affiliation(s)
- Jared C Weddell
- Department of Bioengineering, University of Illinois at Urbana-Champaign, 1304 W Springfield Ave., 3233 Digital Computer Laboratory, Urbana, IL 61801, USA.
| | | |
Collapse
|
22
|
Vélez-Aguilera G, de Dios Gómez-López J, Jiménez-Gutiérrez GE, Vásquez-Limeta A, Laredo-Cisneros MS, Gómez P, Winder SJ, Cisneros B. Control of nuclear β-dystroglycan content is crucial for the maintenance of nuclear envelope integrity and function. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2018; 1865:406-420. [PMID: 29175376 DOI: 10.1016/j.bbamcr.2017.11.013] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Revised: 11/07/2017] [Accepted: 11/20/2017] [Indexed: 01/28/2023]
Abstract
β-Dystroglycan (β-DG) is a plasma membrane protein that has ability to target to the nuclear envelope (NE) to maintain nuclear architecture. Nevertheless, mechanisms controlling β-DG nuclear localization and the physiological consequences of a failure of trafficking are largely unknown. We show that β-DG has a nuclear export pathway in myoblasts that depends on the recognition of a nuclear export signal located in its transmembrane domain, by CRM1. Remarkably, NES mutations forced β-DG nuclear accumulation resulting in mislocalization and decreased levels of emerin and lamin B1 and disruption of various nuclear processes in which emerin (centrosome-nucleus linkage and β-catenin transcriptional activity) and lamin B1 (cell cycle progression and nucleoli structure) are critically involved. In addition to nuclear export, the lifespan of nuclear β-DG is restricted by its nuclear proteasomal degradation. Collectively our data show that control of nuclear β-DG content by the combination of CRM1 nuclear export and nuclear proteasome pathways is physiologically relevant to preserve proper NE structure and activity.
Collapse
Affiliation(s)
- Griselda Vélez-Aguilera
- Departamento de Genética y Biología Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Mexico City, Mexico
| | - Juan de Dios Gómez-López
- Departamento de Genética y Biología Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Mexico City, Mexico
| | - Guadalupe E Jiménez-Gutiérrez
- Departamento de Genética y Biología Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Mexico City, Mexico
| | - Alejandra Vásquez-Limeta
- Departamento de Genética y Biología Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Mexico City, Mexico; Laboratory of Protein Dynamics and Signaling, Center for Cancer Research-Frederick, National Cancer Institute, National Institutes of Health, Frederick, MD 21702, United States
| | - Marco S Laredo-Cisneros
- Departamento de Genética y Biología Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Mexico City, Mexico
| | - Pablo Gómez
- Departamento de Genética y Biología Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Mexico City, Mexico
| | - Steve J Winder
- Department of Biomedical Science, University of Sheffield, Western Bank, Sheffield S10 2TN, United Kingdom
| | - Bulmaro Cisneros
- Departamento de Genética y Biología Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Mexico City, Mexico.
| |
Collapse
|
23
|
Kesten D, Horovitz-Fried M, Brutman-Barazani T, Sampson SR. Insulin-induced translocation of IR to the nucleus in insulin responsive cells requires a nuclear translocation sequence. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2018; 1865:551-559. [PMID: 29317261 DOI: 10.1016/j.bbamcr.2018.01.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Revised: 01/03/2018] [Accepted: 01/04/2018] [Indexed: 02/02/2023]
Abstract
Insulin binding to its cell surface receptor (IR) activates a cascade of events leading to its biological effects. The Insulin-IR complex is rapidly internalized and then is either recycled back to the plasma membrane or sent to lysosomes for degradation. Although most of the receptor is recycled or degraded, a small amount may escape this pathway and migrate to the nucleus of the cell where it might be important in promulgation of receptor signals. In this study we explored the mechanism by which insulin induces IR translocation to the cell nucleus. Experiments were performed cultured L6 myoblasts, AML liver cells and 3T3-L1 adipocytes. Insulin treatment induced a rapid increase in nuclear IR protein levels within 2 to 5 min. Treatment with WGA, an inhibitor of nuclear import, reduced insulin-induced increases nuclear IR protein; IR was, however, translocated to a perinuclear location. Bioinformatics tools predicted a potential nuclear localization sequence (NLS) on IR. Immunofluorescence staining showed that a point mutation on the predicted NLS blocked insulin-induced IR nuclear translocation. In addition, blockade of nuclear IR activation in isolated nuclei by an IR blocking antibody abrogated insulin-induced increases in IR tyrosine phosphorylation and nuclear PKCδ levels. Furthermore, over expression of mutated IR reduced insulin-induced glucose uptake and PKB phosphorylation. When added to isolated nuclei, insulin induced IR phosphorylation but had no effect on nuclear IR protein levels. These results raise questions regarding the possible role of nuclear IR in IR signaling and insulin resistance.
Collapse
Affiliation(s)
- Dov Kesten
- Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan 52900, Israel
| | | | | | - Sanford R Sampson
- Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan 52900, Israel; Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 76100, Israel.
| |
Collapse
|
24
|
Expression and role of TYRO3 and AXL as potential therapeutical targets in leiomyosarcoma. Br J Cancer 2017; 117:1787-1797. [PMID: 29024938 PMCID: PMC5729471 DOI: 10.1038/bjc.2017.354] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Revised: 07/31/2017] [Accepted: 09/07/2017] [Indexed: 12/26/2022] Open
Abstract
Background: Leiomyosarcoma (LMS) are 15% of adult sarcomas and remain seldom curable in metastatic phase. The TAM receptors and their ligands are overexpressed or activated in multiple malignancies, including LMS. Methods: The TAM receptor and ligand expression was evaluated in LMS cell lines and 358 sarcoma samples by either gene expression or immunohistochemistry. TYRO3 and AXL were knocked down. Crizotinib and foretinib were investigated in vitro. Results: High expression of TYRO3 and AXL was detected in LMS cell lines. TYRO3 or AXL gene knockdown reduced cell proliferation/colony formation. Crizotinib and foretinib decreased TYRO3 and AXL phosphorylation, apoptosis, G2/arrest and reduced colony formation. Immunohistochemistry performed in 107 sarcomas showed higher expression of TYRO3 and GAS6 in LMS vs other sarcomas and nuclear TYRO3 only in LMS. Microarray gene expression performed in 251 sarcomas revealed significantly higher expression of TYRO3 and GAS6 in LMS than other sarcomas. Leiomyosarcoma patients with high expression of GAS6 or PROS1 present a significantly worse PFS. Conclusions: Leiomyosarcoma patients, especially those whom develop metastasis, express higher levels of TYRO3 and GAS6. Crizotinib and foretinib showed effective antitumour activity in LMS through TYRO3 and AXL deactivation indicating that clinical trials using TYRO3 and AXL inhibitors are warranted in advanced LMS.
Collapse
|
25
|
Gracida-Jiménez V, Mondragón-González R, Vélez-Aguilera G, Vásquez-Limeta A, Laredo-Cisneros MS, Gómez-López JDD, Vaca L, Gourlay SC, Jacobs LA, Winder SJ, Cisneros B. Retrograde trafficking of β-dystroglycan from the plasma membrane to the nucleus. Sci Rep 2017; 7:9906. [PMID: 28852008 PMCID: PMC5575308 DOI: 10.1038/s41598-017-09972-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Accepted: 07/31/2017] [Indexed: 12/04/2022] Open
Abstract
β-Dystroglycan (β-DG) is a transmembrane protein with critical roles in cell adhesion, cytoskeleton remodeling and nuclear architecture. This functional diversity is attributed to the ability of β-DG to target to, and conform specific protein assemblies at the plasma membrane (PM) and nuclear envelope (NE). Although a classical NLS and importin α/β mediated nuclear import pathway has already been described for β-DG, the intracellular trafficking route by which β-DG reaches the nucleus is unknown. In this study, we demonstrated that β-DG undergoes retrograde intracellular trafficking from the PM to the nucleus via the endosome-ER network. Furthermore, we provided evidence indicating that the translocon complex Sec61 mediates the release of β-DG from the ER membrane, making it accessible for importins and nuclear import. Finally, we show that phosphorylation of β-DG at Tyr890 is a key stimulus for β-DG nuclear translocation. Collectively our data describe the retrograde intracellular trafficking route that β-DG follows from PM to the nucleus. This dual role for a cell adhesion receptor permits the cell to functionally connect the PM with the nucleus and represents to our knowledge the first example of a cell adhesion receptor exhibiting retrograde nuclear trafficking and having dual roles in PM and NE.
Collapse
Affiliation(s)
- Viridiana Gracida-Jiménez
- Departamento de Genética y Biología Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV), Ciudad de México, Mexico, Mexico
| | - Ricardo Mondragón-González
- Departamento de Genética y Biología Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV), Ciudad de México, Mexico, Mexico
| | - Griselda Vélez-Aguilera
- Departamento de Genética y Biología Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV), Ciudad de México, Mexico, Mexico
| | - Alejandra Vásquez-Limeta
- Departamento de Genética y Biología Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV), Ciudad de México, Mexico, Mexico.,Laboratory of Protein Dynamics and Signaling, Center for Cancer Research-Frederick, National Cancer Institute, National Institutes of Health, Frederick, MD, 21702, USA
| | - Marco S Laredo-Cisneros
- Departamento de Genética y Biología Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV), Ciudad de México, Mexico, Mexico
| | - Juan de Dios Gómez-López
- Departamento de Genética y Biología Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV), Ciudad de México, Mexico, Mexico
| | - Luis Vaca
- Instituto de Fisiología Celular, Universidad Nacional Autónoma de Mexico, Ciudad de Mexico, Mexico, Mexico
| | - Sarah C Gourlay
- Department of Biomedical Science, University of Sheffield, Western Bank, Sheffield, S10 2TN, United Kingdom
| | - Laura A Jacobs
- Department of Biomedical Science, University of Sheffield, Western Bank, Sheffield, S10 2TN, United Kingdom
| | - Steve J Winder
- Department of Biomedical Science, University of Sheffield, Western Bank, Sheffield, S10 2TN, United Kingdom
| | - Bulmaro Cisneros
- Departamento de Genética y Biología Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV), Ciudad de México, Mexico, Mexico.
| |
Collapse
|
26
|
Narla ST, Lee YW, Benson C, Sarder P, Brennand K, Stachowiak E, Stachowiak M. Common developmental genome deprogramming in schizophrenia - Role of Integrative Nuclear FGFR1 Signaling (INFS). Schizophr Res 2017; 185:17-32. [PMID: 28094170 PMCID: PMC5507209 DOI: 10.1016/j.schres.2016.12.012] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Revised: 12/06/2016] [Accepted: 12/12/2016] [Indexed: 12/16/2022]
Abstract
The watershed-hypothesis of schizophrenia asserts that over 200 different mutations dysregulate distinct pathways that converge on an unspecified common mechanism(s) that controls disease ontogeny. Consistent with this hypothesis, our RNA-sequencing of neuron committed cells (NCCs) differentiated from established iPSCs of 4 schizophrenia patients and 4 control subjects uncovered a dysregulated transcriptome of 1349 mRNAs common to all patients. Data reveals a global dysregulation of developmental genome, deconstruction of coordinated mRNA networks, and the formation of aberrant, new coordinated mRNA networks indicating a concerted action of the responsible factor(s). Sequencing of miRNA transcriptomes demonstrated an overexpression of 16 miRNAs and deconstruction of interactive miRNA-mRNA networks in schizophrenia NCCs. ChiPseq revealed that the nuclear (n) form of FGFR1, a pan-ontogenic regulator, is overexpressed in schizophrenia NCCs and overtargets dysregulated mRNA and miRNA genes. The nFGFR1 targeted 54% of all human gene promoters and 84.4% of schizophrenia dysregulated genes. The upregulated genes reside within major developmental pathways that control neurogenesis and neuron formation, whereas downregulated genes are involved in oligodendrogenesis. Our results indicate (i) an early (preneuronal) genomic etiology of schizophrenia, (ii) dysregulated genes and new coordinated gene networks are common to unrelated cases of schizophrenia, (iii) gene dysregulations are accompanied by increased nFGFR1-genome interactions, and (iv) modeling of increased nFGFR1 by an overexpression of a nFGFR1 lead to up or downregulation of selected genes as observed in schizophrenia NCCs. Together our results designate nFGFR1 signaling as a potential common dysregulated mechanism in investigated patients and potential therapeutic target in schizophrenia.
Collapse
Affiliation(s)
- S. T. Narla
- Department of Pathology and Anatomical Sciences, State University of New York at Buffalo, Buffalo, NY, USA,Western New York Stem Cell Culture and Analysis Center, State University of New York at Buffalo, Buffalo, NY, USA
| | - Y-W. Lee
- Department of Pathology and Anatomical Sciences, State University of New York at Buffalo, Buffalo, NY, USA
| | - C.A. Benson
- Department of Pathology and Anatomical Sciences, State University of New York at Buffalo, Buffalo, NY, USA,Western New York Stem Cell Culture and Analysis Center, State University of New York at Buffalo, Buffalo, NY, USA
| | - P. Sarder
- Department of Pathology and Anatomical Sciences, State University of New York at Buffalo, Buffalo, NY, USA
| | - K. Brennand
- Icahn School of Medicine at Mount Sinai, Departments of Psychiatry and Neuroscience, New York, NY, USA
| | - E.K. Stachowiak
- Department of Pathology and Anatomical Sciences, State University of New York at Buffalo, Buffalo, NY, USA,Western New York Stem Cell Culture and Analysis Center, State University of New York at Buffalo, Buffalo, NY, USA
| | - M.K. Stachowiak
- Department of Pathology and Anatomical Sciences, State University of New York at Buffalo, Buffalo, NY, USA,Western New York Stem Cell Culture and Analysis Center, State University of New York at Buffalo, Buffalo, NY, USA,Correspondence should be addressed to Michal K. Stachowiak Department of Pathology and Anatomical Sciences, SUNY, 3435 Main Street, 206A Farber Hall, Buffalo, N.Y. 14214, tel. (716) 829 3540
| |
Collapse
|
27
|
Fu J, Xia A, Qi X. Identification of novel peptoid agonists of fibroblast growth factor receptor using microarray-based screening. MEDCHEMCOMM 2016; 7:1183-1189. [PMID: 27721968 DOI: 10.1039/c6md00121a] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Drug development targeting fibroblast growth factor receptors (FGFRs) represents an emerging theme in the field of medicinal chemistry. Considering the fact that most of the currently identified FGFR agonists are long chain peptides with limited stability, the discovery of novel non-peptide FGFR ligands is still highly demanded. A linear one-bead-one-compound peptoid (oligomers of N-substituted glycine units) library with a theoretical diversity of 106 was designed and synthesized. Microarray-based screening led to the identification of four hit sequences 1-4 as FGFR1α ligands, which were further confirmed using both solution-phase and solid-phase binding assays. Western blot results indicated that peptoids 2-4 activated FGFR signaling pathways, resulting in increased levels of p-Akt and p-ERK in different cell lines. Our work discovered novel peptoid ligands as FGFR agonists, shedding new light on FGFR-based drug discovery.
Collapse
Affiliation(s)
- Junjie Fu
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, FL, 32610
| | - Amy Xia
- Columbia University, New York, NY 10027
| | - Xin Qi
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, FL, 32610
| |
Collapse
|
28
|
Heck D, Wortmann S, Kraus L, Ronchi CL, Sinnott RO, Fassnacht M, Sbiera S. Role of Endocrine Gland-Derived Vascular Endothelial Growth Factor (EG-VEGF) and Its Receptors in Adrenocortical Tumors. Discov Oncol 2016; 6:225-36. [PMID: 26475302 PMCID: PMC4630243 DOI: 10.1007/s12672-015-0236-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2015] [Accepted: 10/02/2015] [Indexed: 12/19/2022] Open
Abstract
Angiogenesis is essential for tumor growth and metastasis. Endocrine gland-derived vascular endothelial growth factor (EG-VEGF) is an angiogenic factor predominantly expressed in steroidogenic organs like the adrenal gland, ovary, testes, and placenta. EG-VEGF has antiapoptotic, mitogenic, and chemoattractive properties mediated via the two G protein-coupled receptors prokineticin receptor 1 (PKR1) and prokineticin receptor 2 (PKR2). We investigated the expression of EG-VEGF and its receptors in a large number of normal adrenal glands (NAG), adrenocortical adenomas (ACA), and carcinomas (ACC) using real-time PCR (NAG, n = 12; ACA, n = 24; and ACC, n = 30) and immunohistochemistry (NAG, n = 9; ACA, n = 23; and ACC, n = 163) and evaluated its impact on patients’ survival. EG-VEGF, PKR1, and PKR2 mRNA and protein are expressed in NAG and the vast majority of ACA and ACC samples. The mean EG-VEGF mRNA expression was significantly lower in ACC (606.5 ± 77.1 copies) compared to NAG (4,043 ± 1,111) and cortisol-producing adenomas (CPA) (4,433 ± 2,378) (p < 0.01 and p < 0.05, respectively). However, cytoplasmic and nuclear EG-VEGF protein expression was either significantly higher or similar in ACC (H score 2.4 ± 0.05, p < 0.05 and 1.7 ± 0.08, n.s., respectively) compared to NAG (1.8 ± 0.14 and 1.7 ± 0.2). Nuclear protein expression of either EG-VEGF or PKR1 or both is predictive for a higher mortality compared to patients without nuclear expression (hazard ratio (HR) = 5.15; 95 % confidence interval (CI) = 1.24–21.36, n = 100, p = 0.02 independent of age, sex, and tumor stage). These findings suggest that EG-VEGF and its receptor PKR1 might play a role in the pathogenesis of adrenocortical tumors and could serve as prognostic markers for this rare malignant disease.
Collapse
|
29
|
Rosa ACG, Soares AB, Santos FP, Furuse C, de Araújo VC. Immunoexpression of growth factors and receptors in polymorphous low-grade adenocarcinoma. J Oral Pathol Med 2015; 45:494-9. [PMID: 26671844 DOI: 10.1111/jop.12402] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/18/2015] [Indexed: 11/30/2022]
Abstract
BACKGROUND Polymorphous low-grade adenocarcinoma (PLGA) is a rare malignant tumor that usually arises in the minor salivary glands. Growth factors are cell-secreted peptides that regulate biological processes such as growth, proliferation, and differentiation. In salivary gland tumors, immunoexpression of growth factors and their receptors is associated with cell proliferation, malignant transformation, and tumor invasion. This study analyzed the expression of growth factors and receptors in PLGA, in other to better understand the mechanisms involved in the process of neoplastic cell proliferation and tumor progression. METHODS The expression of growth factors FGF-2, PDGF-A, PDGF-B and receptors FGFR-1, FGFR-2, PDGFR-α, and EGFR was analyzed in 24 PLGA samples in comparison with normal salivary glands, by immunohistochemistry. A semi-quantitative analysis determined cell positivity in all stained sections. Scores were assigned according to percentage of reactive cells: score 0 < 10%; score 1-10 to 25%; score 2-25% to 50%; score 3->50%. The level of significance was set at 5%. RESULTS Most of the growth factors and receptors, apart from FGFR-2, were significantly reactive in PLGA. Comparing to salivary acini, all of the reactive growth factors and receptors were significantly stronger in PLGA. Comparing to salivary ducts, the expression of FGF-2, PDGF-B, FGFR-1, and EGFR was significantly stronger in the nuclei and/or cytoplasm of the neoplastic cells. CONCLUSIONS The increased expression of the growth factors and receptors in the PLGA, compared to normal salivary glands, may be related to cell proliferation, somehow participating in the oncogenic process.
Collapse
Affiliation(s)
- Ana Cláudia Garcia Rosa
- Department of Oral Pathology, Lutheran University of Palmas, Tocantins, Brazil.,Department of Health Sciences, Federal University of Tocantins, Palmas, Tocantins, Brazil
| | - Andresa Borges Soares
- Department of Oral Pathology, São Leopoldo Mandic Institute and Research Center, Campinas, São Paulo, Brazil
| | - Fabrício Passador Santos
- Department of Oral Pathology, São Leopoldo Mandic Institute and Research Center, Campinas, São Paulo, Brazil
| | - Cristiane Furuse
- Department of Oral Pathology, São Paulo State University, Araçatuba, São Paulo, Brazil
| | - Vera Cavalcanti de Araújo
- Department of Oral Pathology, São Leopoldo Mandic Institute and Research Center, Campinas, São Paulo, Brazil
| |
Collapse
|
30
|
Saucedo L, Buffa GN, Rosso M, Guillardoy T, Góngora A, Munuce MJ, Vazquez-Levin MH, Marín-Briggiler C. Fibroblast Growth Factor Receptors (FGFRs) in Human Sperm: Expression, Functionality and Involvement in Motility Regulation. PLoS One 2015; 10:e0127297. [PMID: 25970615 PMCID: PMC4430232 DOI: 10.1371/journal.pone.0127297] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2014] [Accepted: 04/13/2015] [Indexed: 11/18/2022] Open
Abstract
Fibroblast growth factors receptors (FGFRs) have been widely characterized in somatic cells, but there is scarce evidence of their expression and function in mammalian gametes. The objective of the present study was to evaluate the expression of FGFRs in human male germ cells, to determine sperm FGFR activation by the FGF2 ligand and their participation in the regulation of sperm motility. The expression of FGFR1, 2, 3 and 4 mRNAs and proteins in human testis and localization of these receptors in germ cells of the seminiferous epithelium was demonstrated. In ejaculated sperm, FGFRs were localized to the acrosomal region and flagellum. Sperm exposure to FGF2 caused an increase in flagellar FGFR phosphorylation and activation of extracellular signal-regulated kinase (ERK) and protein kinase B (PKB or Akt) signaling pathways. Incubation with FGF2 led to a significant increase in the percentage of total and progressive sperm motility, as well as in sperm kinematics. All responses were prevented by sperm preincubation with BGJ398, a specific inhibitor of FGFR tyrosine kinase activity. In addition to confirming the expression of FGFRs in germ cells of the human testis, our study describes for the first time the presence, localization and functionality of human sperm FGFRs, and provides evidence of the beneficial effect of FGF2 upon sperm motility.
Collapse
Affiliation(s)
- Lucía Saucedo
- Instituto de Biología y Medicina Experimental (IBYME), CONICET-FIBYME, Buenos Aires, Argentina
| | - Gabriela N. Buffa
- Instituto de Biología y Medicina Experimental (IBYME), CONICET-FIBYME, Buenos Aires, Argentina
| | - Marina Rosso
- Instituto de Biología y Medicina Experimental (IBYME), CONICET-FIBYME, Buenos Aires, Argentina
| | - Tomás Guillardoy
- Instituto de Biología y Medicina Experimental (IBYME), CONICET-FIBYME, Buenos Aires, Argentina
| | - Adrian Góngora
- Instituto de Biología y Medicina Experimental (IBYME), CONICET-FIBYME, Buenos Aires, Argentina
| | - María J. Munuce
- Laboratorio de Medicina Reproductiva, Area de Bioquímica Clínica, Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, Rosario, Santa Fe, Argentina
- Reprolab, Sanatorio Británico de Rosario, Rosario, Santa Fe, Argentina
| | - Mónica H. Vazquez-Levin
- Instituto de Biología y Medicina Experimental (IBYME), CONICET-FIBYME, Buenos Aires, Argentina
| | - Clara Marín-Briggiler
- Instituto de Biología y Medicina Experimental (IBYME), CONICET-FIBYME, Buenos Aires, Argentina
- * E-mail:
| |
Collapse
|
31
|
Förthmann B, Grothe C, Claus P. A nuclear odyssey: fibroblast growth factor-2 (FGF-2) as a regulator of nuclear homeostasis in the nervous system. Cell Mol Life Sci 2015; 72:1651-62. [PMID: 25552245 PMCID: PMC11113852 DOI: 10.1007/s00018-014-1818-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2014] [Revised: 12/10/2014] [Accepted: 12/19/2014] [Indexed: 01/07/2023]
Abstract
Nuclear localization of classical growth factors is a well-known phenomenon but still remains a molecular and cellular conundrum. Fibroblast growth factor-2 (FGF-2) is an excellent example of a protein which functions as an extracellular molecule involved in canonical receptor tyrosine kinase signaling as well as displaying intracellular functions. Paracrine and nuclear functions are two important sides of the same protein. FGF-2 is expressed in isoforms with different molecular weights from one mRNA species. In rodents, all of these isoforms become imported to the nucleus. In this review, we discuss structural and functional aspects of FGF-2 isoforms in the nervous system. The nuclear odyssey of FGF-2 is reflected by nuclear dynamics, localization to nuclear bodies such as nucleoli, binding to chromatin and engagement in various protein interactions. Recently discovered molecular partnerships of the isoforms shed light on their nuclear functions, thereby greatly extending our knowledge of the multifaceted functions of FGF-2.
Collapse
Affiliation(s)
- Benjamin Förthmann
- Department of Neuroanatomy, Institute of Neuroanatomy, Hannover Medical School, OE 4140, Carl-Neuberg-Str.1, 30625 Hannover, Germany
| | - Claudia Grothe
- Department of Neuroanatomy, Institute of Neuroanatomy, Hannover Medical School, OE 4140, Carl-Neuberg-Str.1, 30625 Hannover, Germany
- Center for Systems Neuroscience, 30625 Hannover, Germany
| | - Peter Claus
- Department of Neuroanatomy, Institute of Neuroanatomy, Hannover Medical School, OE 4140, Carl-Neuberg-Str.1, 30625 Hannover, Germany
- Center for Systems Neuroscience, 30625 Hannover, Germany
| |
Collapse
|
32
|
Ferreira MM, Dewi RE, Heilshorn SC. Microfluidic analysis of extracellular matrix-bFGF crosstalk on primary human myoblast chemoproliferation, chemokinesis, and chemotaxis. Integr Biol (Camb) 2015; 7:569-79. [PMID: 25909157 PMCID: PMC4528978 DOI: 10.1039/c5ib00060b] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Exposing myoblasts to basic fibroblast growth factor (bFGF), which is released after muscle injury, results in receptor phosphorylation, faster migration, and increased proliferation. These effects occur on time scales that extend across three orders of magnitude (10(0)-10(3) minutes). Finite element modeling of Transwell assays, which are traditionally used to assess chemotaxis, revealed that the bFGF gradient formed across the membrane pore is short-lived and diminishes 45% within the first minute. Thus, to evaluate bFGF-induced migration over 10(2) minutes, we employed a microfluidic assay capable of producing a stable, linear concentration gradient to perform single-cell analyses of chemokinesis and chemotaxis. We hypothesized that the composition of the underlying extracellular matrix (ECM) may affect the behavioral response of myoblasts to soluble bFGF, as previous work with other cell types has suggested crosstalk between integrin and fibroblast growth factor (FGF) receptors. Consistent with this notion, we found that bFGF significantly reduced the doubling time of myoblasts cultured on laminin but not fibronectin or collagen. Laminin also promoted significantly faster migration speeds (13.4 μm h(-1)) than either fibronectin (10.6 μm h(-1)) or collagen (7.6 μm h(-1)) without bFGF stimulation. Chemokinesis driven by bFGF further increased migration speed in a strictly additive manner, resulting in an average increase of 2.3 μm h(-1) across all ECMs tested. We observed relatively mild chemoattraction (∼67% of myoblast population) in response to bFGF gradients of 3.2 ng mL(-1) mm(-1) regardless of ECM identity. Thus, while ECM-bFGF crosstalk did impact chemoproliferation, it did not have a significant effect on chemokinesis or chemotaxis. These data suggest that the main physiological effect of bFGF on myoblast migration is chemokinesis and that changes in the surrounding ECM, resulting from aging and/or disease may impact muscle regeneration by altering myoblast migration and proliferation.
Collapse
Affiliation(s)
| | - Ruby E. Dewi
- Department of Materials Science and Engineering, Stanford University
| | | |
Collapse
|
33
|
Terranova C, Narla ST, Lee YW, Bard J, Parikh A, Stachowiak EK, Tzanakakis ES, Buck MJ, Birkaya B, Stachowiak MK. Global Developmental Gene Programing Involves a Nuclear Form of Fibroblast Growth Factor Receptor-1 (FGFR1). PLoS One 2015; 10:e0123380. [PMID: 25923916 PMCID: PMC4414453 DOI: 10.1371/journal.pone.0123380] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Accepted: 02/17/2015] [Indexed: 12/11/2022] Open
Abstract
Genetic studies have placed the Fgfr1 gene at the top of major ontogenic pathways that enable gastrulation, tissue development and organogenesis. Using genome-wide sequencing and loss and gain of function experiments the present investigation reveals a mechanism that underlies global and direct gene regulation by the nuclear form of FGFR1, ensuring that pluripotent Embryonic Stem Cells differentiate into Neuronal Cells in response to Retinoic Acid. Nuclear FGFR1, both alone and with its partner nuclear receptors RXR and Nur77, targets thousands of active genes and controls the expression of pluripotency, homeobox, neuronal and mesodermal genes. Nuclear FGFR1 targets genes in developmental pathways represented by Wnt/β-catenin, CREB, BMP, the cell cycle and cancer-related TP53 pathway, neuroectodermal and mesodermal programing networks, axonal growth and synaptic plasticity pathways. Nuclear FGFR1 targets the consensus sequences of transcription factors known to engage CREB-binding protein, a common coregulator of transcription and established binding partner of nuclear FGFR1. This investigation reveals the role of nuclear FGFR1 as a global genomic programmer of cell, neural and muscle development.
Collapse
Affiliation(s)
- Christopher Terranova
- Department of Pathology and Anatomical Sciences, Western New York Stem Cell Culture and Analysis Center, State University of New York at Buffalo, Buffalo, New York, United States of America
| | - Sridhar T. Narla
- Department of Pathology and Anatomical Sciences, Western New York Stem Cell Culture and Analysis Center, State University of New York at Buffalo, Buffalo, New York, United States of America
| | - Yu-Wei Lee
- Department of Pathology and Anatomical Sciences, Western New York Stem Cell Culture and Analysis Center, State University of New York at Buffalo, Buffalo, New York, United States of America
| | - Jonathan Bard
- Next-Generation Sequencing and Expression Analysis Core, State University of New York at Buffalo, Buffalo, New York, United States of America
| | - Abhirath Parikh
- Department of Chemical and Biological Engineering, Western New York Stem Cell Culture and Analysis Center, State University of New York at Buffalo, Buffalo, New York, United States of America
| | - Ewa K. Stachowiak
- Department of Pathology and Anatomical Sciences, Western New York Stem Cell Culture and Analysis Center, State University of New York at Buffalo, Buffalo, New York, United States of America
| | - Emmanuel S. Tzanakakis
- Department of Chemical and Biological Engineering, Western New York Stem Cell Culture and Analysis Center, State University of New York at Buffalo, Buffalo, New York, United States of America
| | - Michael J. Buck
- Department of Biochemistry, Genomics and Bioinformatics Core, Western New York Stem Cell Culture and Analysis Center, State University of New York at Buffalo, Buffalo, New York, United States of America
| | - Barbara Birkaya
- Department of Pathology and Anatomical Sciences, Western New York Stem Cell Culture and Analysis Center, State University of New York at Buffalo, Buffalo, New York, United States of America
| | - Michal K. Stachowiak
- Department of Pathology and Anatomical Sciences, Western New York Stem Cell Culture and Analysis Center, State University of New York at Buffalo, Buffalo, New York, United States of America
- * E-mail:
| |
Collapse
|
34
|
Stachowiak MK, Birkaya B, Aletta JM, Narla ST, Benson CA, Decker B, Stachowiak EK. "Nuclear FGF receptor-1 and CREB binding protein: an integrative signaling module". J Cell Physiol 2015; 230:989-1002. [PMID: 25503065 DOI: 10.1002/jcp.24879] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2014] [Accepted: 12/05/2014] [Indexed: 12/15/2022]
Abstract
In this review we summarize the current understanding of a novel integrative function of Fibroblast Growth Factor Receptor-1 (FGFR1) and its partner CREB Binding Protein (CBP) acting as a nuclear regulatory complex. Nuclear FGFR1 and CBP interact with and regulate numerous genes on various chromosomes. FGFR1 dynamic oscillatory interactions with chromatin and with specific genes, underwrites gene regulation mediated by diverse developmental signals. Integrative Nuclear FGFR1 Signaling (INFS) effects the differentiation of stem cells and neural progenitor cells via the gene-controlling Feed-Forward-And-Gate mechanism. Nuclear accumulation of FGFR1 occurs in numerous cell types and disruption of INFS may play an important role in developmental disorders such as schizophrenia, and in metastatic diseases such as cancer. Enhancement of INFS may be used to coordinate the gene regulation needed to activate cell differentiation for regenerative purposes or to provide interruption of cancer stem cell proliferation.
Collapse
Affiliation(s)
- Michal K Stachowiak
- Department of Pathology and Anatomical Sciences, Western New York Stem Cells Culture and Analysis Center, State University of New York, Buffalo
| | | | | | | | | | | | | |
Collapse
|
35
|
Miklos Z, Flynn FW, Lessard A. Stress-induced dendritic internalization and nuclear translocation of the neurokinin-3 (NK3) receptor in vasopressinergic profiles of the rat paraventricular nucleus of the hypothalamus. Brain Res 2014; 1590:31-44. [DOI: 10.1016/j.brainres.2014.09.043] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2014] [Revised: 09/16/2014] [Accepted: 09/18/2014] [Indexed: 01/31/2023]
|
36
|
A putative pH-dependent nuclear localization signal in the juxtamembrane region of c-Met. Exp Mol Med 2014; 46:e119. [PMID: 25341359 PMCID: PMC4221695 DOI: 10.1038/emm.2014.67] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2014] [Revised: 09/05/2014] [Accepted: 09/10/2014] [Indexed: 12/20/2022] Open
Abstract
The C-terminal fragment of the c-Met receptor tyrosine kinase is present in the nuclei of some cells irrespective of ligand stimulation, but the responsible nuclear localization signal (NLS) has not been previously reported. Here, we report that two histidine residues separated by a 10-amino-acid spacer (H1068-H1079) located in the juxtamembrane region of c-Met function as a putative novel NLS. Deletion of these sequences significantly abolished the nuclear translocation of c-Met, as did substitution of the histidines with alanines. This substitution also decreased the association of c-Met fragment with importin β. The putative NLS of c-Met is unique in that it relies on histidines, whose positive charge changes depending on pH, rather than the lysines or arginines, commonly found in classical bipartite NLSs, suggesting the possible 'pH-dependency' of this NLS. Indeed, decreasing the cytosolic pH either with nigericin, an Na(+)/H(+) exchanger or pH 6.5 KRB buffer significantly increased the level of nuclear c-Met and the interaction of the c-Met fragment with importin β, indicating that low pH itself enhanced nuclear translocation. Consistent with this, nigericin treatment also increased the nuclear level of endogenous c-Met in HeLa cells. The putative aberrant bipartite NLS of c-Met seems to be the first example of what we call a 'pH-dependent' NLS.
Collapse
|
37
|
Lewerenz J, Baxter P, Kassubek R, Albrecht P, Van Liefferinge J, Westhoff MA, Halatsch ME, Karpel-Massler G, Meakin PJ, Hayes JD, Aronica E, Smolders I, Ludolph AC, Methner A, Conrad M, Massie A, Hardingham GE, Maher P. Phosphoinositide 3-kinases upregulate system xc(-) via eukaryotic initiation factor 2α and activating transcription factor 4 - A pathway active in glioblastomas and epilepsy. Antioxid Redox Signal 2014; 20:2907-22. [PMID: 24219064 PMCID: PMC4038988 DOI: 10.1089/ars.2013.5455] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2013] [Revised: 10/25/2013] [Accepted: 11/12/2013] [Indexed: 01/08/2023]
Abstract
AIMS Phosphoinositide 3-kinases (PI3Ks) relay growth factor signaling and mediate cytoprotection and cell growth. The cystine/glutamate antiporter system xc(-) imports cystine while exporting glutamate, thereby promoting glutathione synthesis while increasing extracellular cerebral glutamate. The aim of this study was to analyze the pathway through which growth factor and PI3K signaling induce the cystine/glutamate antiporter system xc(-) and to demonstrate its biological significance for neuroprotection, cell growth, and epilepsy. RESULTS PI3Ks induce system xc(-) through glycogen synthase kinase 3β (GSK-3β) inhibition, general control non-derepressible-2-mediated eukaryotic initiation factor 2α phosphorylation, and the subsequent translational up-regulation of activating transcription factor 4. This pathway is essential for PI3Ks to modulate oxidative stress resistance of nerve cells and insulin-induced growth in fibroblasts. Moreover, the pathway is active in human glioblastoma cells. In addition, it is induced in primary cortical neurons in response to robust neuronal activity and in hippocampi from patients with temporal lobe epilepsy. INNOVATION Our findings further extend the concepts of how growth factors and PI3Ks induce neuroprotection and cell growth by adding a new branch to the signaling network downstream of GSK-3β, which, ultimately, leads to the induction of the cystine/glutamate antiporter system xc(-). Importantly, the induction of this pathway by neuronal activity and in epileptic hippocampi points to a potential role in epilepsy. CONCLUSION PI3K-regulated system xc(-) activity is not only involved in the stress resistance of neuronal cells and in cell growth by increasing the cysteine supply and glutathione synthesis, but also plays a role in the pathophysiology of tumor- and non-tumor-associated epilepsy by up-regulating extracellular cerebral glutamate.
Collapse
Affiliation(s)
- Jan Lewerenz
- Department of Neurology, Ulm University, Ulm, Germany
| | - Paul Baxter
- Center for Integrative Physiology, University of Edinburgh, Edinburgh, Scotland, United Kingdom
| | | | - Philipp Albrecht
- Department of Neurology, Medical Faculty, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Joeri Van Liefferinge
- Pharmaceutical Chemistry and Drug Analysis, Center for Neurosciences, Vrije Universiteit Brussel, Brussels, Belgium
| | | | | | | | - Paul J. Meakin
- Medical Research Institute, University of Dundee, Ninewells Hospital and Medical School, Dundee, Scotland, United Kingdom
| | - John D. Hayes
- Medical Research Institute, University of Dundee, Ninewells Hospital and Medical School, Dundee, Scotland, United Kingdom
| | - Eleonora Aronica
- Department of (Neuro)Pathology, Academic Medical Center, and Swammerdam Institute for Life Sciences, Center for Neuroscience, University of Amsterdam, Amsterdam, The Netherlands
- SEIN—Stichting Epilepsie Instellingen Nederland, Heemstede, The Netherlands
| | - Ilse Smolders
- Pharmaceutical Chemistry and Drug Analysis, Center for Neurosciences, Vrije Universiteit Brussel, Brussels, Belgium
| | | | - Axel Methner
- Focus Program Translational Neuroscience (FTN), Rhine Main Neuroscience Network (rmn), Department of Neurology, Johannes Gutenberg University Medical Center Mainz, Mainz, Germany
| | - Marcus Conrad
- Institute of Developmental Genetics, Helmholtz Zentrum München, Neuherberg, Germany
| | - Ann Massie
- Pharmaceutical Biotechnology and Molecular Biology, Center for Neurosciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Giles E. Hardingham
- Center for Integrative Physiology, University of Edinburgh, Edinburgh, Scotland, United Kingdom
| | - Pamela Maher
- Cellular Neurobiology Laboratory, Salk Institute for Biological Studies, La Jolla, California
| |
Collapse
|
38
|
Garcia-Dorado D, Ruiz-Meana M, Rodríguez-Sinovas A. Connexin 43 phosphorylation in subsarcolemmal mitochondria: a general cardioprotective signal targeted by fibroblast growth factor-2? Cardiovasc Res 2014; 103:1-2. [PMID: 24835275 DOI: 10.1093/cvr/cvu129] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Affiliation(s)
- David Garcia-Dorado
- Laboratory of Experimental Cardiology, Cardiology Department, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, Pg. Vall d'Hebron 119-129, Barcelona 08035, Spain
| | - Marisol Ruiz-Meana
- Laboratory of Experimental Cardiology, Cardiology Department, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, Pg. Vall d'Hebron 119-129, Barcelona 08035, Spain
| | - Antonio Rodríguez-Sinovas
- Laboratory of Experimental Cardiology, Cardiology Department, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, Pg. Vall d'Hebron 119-129, Barcelona 08035, Spain
| |
Collapse
|
39
|
Kovalszky I, Hjerpe A, Dobra K. Nuclear translocation of heparan sulfate proteoglycans and their functional significance. Biochim Biophys Acta Gen Subj 2014; 1840:2491-7. [PMID: 24780644 DOI: 10.1016/j.bbagen.2014.04.015] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2013] [Revised: 04/17/2014] [Accepted: 04/18/2014] [Indexed: 12/25/2022]
Abstract
BACKGROUND Heparan sulfate proteoglycans (HSPGs) are important constituents of the cell membrane and they act as co-receptors for cellular signaling. Syndecan-1, glypican and perlecan also translocate to the nucleus in a regulated manner. Similar nuclear transport of growth factors and heparanase indicate a possible co-regulation and functional significance. SCOPE OF REVIEW In this review we dissect the structural requirement for the nuclear translocation of HSPGs and their functional implications.s MAJOR CONCLUSIONS The functions of the nuclear HSPGs are still incompletely understood. Evidence point to possible functions in hampering cell proliferation, inhibition of DNA topoisomerase I activity and inhibition of gene transcription. GENERAL SIGNIFICANCE HSPGs influence the behavior of malignant tumors in many different ways. Modulating their functions may offer powerful tools to control fundamental biological processes and provide the basis for subsequent targeted therapies in cancer. This article is part of a Special Issue entitled Matrix-mediated cell behaviour and properties.
Collapse
Affiliation(s)
- Ilona Kovalszky
- First Department of Pathology & Experimental Cancer Research Semmelweis University, Üllői street 26, Budapest 1085, Hungary
| | - Anders Hjerpe
- Department of Laboratory Medicine, Karolinska Institutet, Karolinska University Hospital F46, SE-141 86 Stockholm Sweden
| | - Katalin Dobra
- Department of Laboratory Medicine, Karolinska Institutet, Karolinska University Hospital F46, SE-141 86 Stockholm Sweden.
| |
Collapse
|
40
|
Abstract
FGFR (fibroblast growth factor receptor) signalling plays critical roles in embryogensis, adult physiology, tissue repair and many pathologies. Of particular interest over recent years, it has been implicated in a wide range of cancers, and concerted efforts are underway to target different aspects of FGFR signalling networks. A major focus has been identifying the canonical downstream signalling pathways in cancer cells, and these are now relatively well understood. In the present review, we focus on two distinct but emerging hot topics in FGF biology: its role in stromal cross-talk during cancer progression and the potential roles of FGFR signalling in the nucleus. These neglected areas are proving to be of great interest clinically and are intimately linked, at least in pancreatic cancer. The importance of the stroma in cancer is well accepted, both as a conduit/barrier for treatment and as a target in its own right. Nuclear receptors are less acknowledged as targets, largely due to historical scepticism as to their existence or importance. However, increasing evidence from across the receptor tyrosine kinase field is now strong enough to make the study of nuclear growth factor receptors a major area of interest.
Collapse
|
41
|
Burgazli KM, Behrendt MA, Mericliler M, Chasan R, Parahuleva M, Erdogan A. The impact of statins on FGF-2-stimulated human umbilical vein endothelial cells. Postgrad Med 2014; 126:118-28. [PMID: 24393759 DOI: 10.3810/pgm.2014.01.2732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
AIM To determine the effects of different types of statins on proliferative and migrative behaviors of basic fibroblastic growth factor (FGF)-2-stimulated endothelial cells. MATERIALS AND METHODS Human umbilical vein endothelial cells (HUVECs) were isolated and cultured. Groups were arranged in order to observe the impact of each individual substance alone, or under stimulation with statin on FGF-2-stimulated endothelial cells. Endothelial cells were stimulated with human growth factor (HGF), statins, methyl-β-cyclodextrin (β-MCD), and either farnesyl pyrophosphate (FPP) ammonium salt, or geranylgeranyl-pyrophosphate (GGPP), respectively. Cell proliferation analyses were performed 48 hours after stimulation and gaps between migration borders were used in migration analyses. RESULTS The statins showed significant antiproliferative and anti-migrative effects and inhibited the proliferative behavior of FGF-2. However, endothelial cell proliferation and migration were significantly increased after mevalonate co-incubation. Experiments with β-MCD indicated that the destruction of lipid rafts had a negative impact on the action of FGF-2. Stimulation of statin-incubated cells with FPP had no additional effect on proliferation or migration. Notably, although FGF-2 exerted a pro-migrative effect, the effect was not shown in the FPP + FGF-2 group. The anti-migrative actions of statins along with disruption of membrane integrity were reversed by the addition of GGPP. CONCLUSION The angiogenic effect of FGF-2 is suppressed through inhibition of the intracellular cholesterol biosynthesis via statins. Inhibitory effects of statins on FGF-2-stimulated HUVECs were observed to result from both the inhibition of isoprenylation and the destruction of lipid rafts on the cell membrane.
Collapse
Affiliation(s)
- Kamil Mehmet Burgazli
- Department of Internal Medicine and Angiology, Wuppertal Research and Medical Center, Wuppertal, Germany.
| | | | | | | | | | | |
Collapse
|
42
|
Jahanshahi A, Schönfeld LM, Lemmens E, Hendrix S, Temel Y. In vitro and in vivo neuronal electrotaxis: a potential mechanism for restoration? Mol Neurobiol 2013; 49:1005-16. [PMID: 24243342 DOI: 10.1007/s12035-013-8575-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2013] [Accepted: 10/21/2013] [Indexed: 01/19/2023]
Abstract
Electrical brain stimulation used to treat a variety of neurological and psychiatric diseases is entering a new period. The technique is well established and the potential complications are well known and generally manageable. Recent studies demonstrated that electrical fields (EFs) can enhance neuroplasticity-related processes. EFs applied in the physiological range induce migration of different neural cell types from different species in vitro. There are some evidences that also the speed and directedness of cell migration are enhanced by EFs. However, it is still unclear how electrical signals from the extracellular space are translated into intracellular actions resulting in the so-called electrotaxis phenomenon. Here, we aim to provide a comprehensive review of the data on responses of cells to electrical stimulation and the relation to functional recovery.
Collapse
Affiliation(s)
- Ali Jahanshahi
- Department of Neuroscience, Maastricht University Medical Center, Maastricht, the Netherlands,
| | | | | | | | | |
Collapse
|
43
|
Stachowiak MK, Kucinski A, Curl R, Syposs C, Yang Y, Narla S, Terranova C, Prokop D, Klejbor I, Bencherif M, Birkaya B, Corso T, Parikh A, Tzanakakis ES, Wersinger S, Stachowiak EK. Schizophrenia: a neurodevelopmental disorder--integrative genomic hypothesis and therapeutic implications from a transgenic mouse model. Schizophr Res 2013; 143:367-76. [PMID: 23231877 DOI: 10.1016/j.schres.2012.11.004] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2012] [Revised: 11/02/2012] [Accepted: 11/06/2012] [Indexed: 12/14/2022]
Abstract
Schizophrenia is a neurodevelopmental disorder featuring complex aberrations in the structure, wiring, and chemistry of multiple neuronal systems. The abnormal developmental trajectory of the brain appears to be established during gestation, long before clinical symptoms of the disease appear in early adult life. Many genes are associated with schizophrenia, however, altered expression of no one gene has been shown to be present in a majority of schizophrenia patients. How does altered expression of such a variety of genes lead to the complex set of abnormalities observed in the schizophrenic brain? We hypothesize that the protein products of these genes converge on common neurodevelopmental pathways that affect the development of multiple neural circuits and neurotransmitter systems. One such neurodevelopmental pathway is Integrative Nuclear FGFR1 Signaling (INFS). INFS integrates diverse neurogenic signals that direct the postmitotic development of embryonic stem cells, neural progenitors and immature neurons, by direct gene reprogramming. Additionally, FGFR1 and its partner proteins link multiple upstream pathways in which schizophrenia-linked genes are known to function and interact directly with those genes. A th-fgfr1(tk-) transgenic mouse with impaired FGF receptor signaling establishes a number of important characteristics that mimic human schizophrenia - a neurodevelopmental origin, anatomical abnormalities at birth, a delayed onset of behavioral symptoms, deficits across multiple domains of the disorder and symptom improvement with typical and atypical antipsychotics, 5-HT antagonists, and nicotinic receptor agonists. Our research suggests that altered FGF receptor signaling plays a central role in the developmental abnormalities underlying schizophrenia and that nicotinic agonists are an effective class of compounds for the treatment of schizophrenia.
Collapse
Affiliation(s)
- M K Stachowiak
- Molecular and Structural Neurobiology & Gene Therapy Program, Department of Pathology and Anatomical Sciences, Western New York Stem Cell Culture and Analysis Center, SUNY, Buffalo, NY, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Chioni AM, Grose R. FGFR1 cleavage and nuclear translocation regulates breast cancer cell behavior. ACTA ACUST UNITED AC 2012; 197:801-17. [PMID: 22665522 PMCID: PMC3373409 DOI: 10.1083/jcb.201108077] [Citation(s) in RCA: 95] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
FGFR1 cleavage by Granzyme B induces its nuclear translocation, in which it stimulates cell migration through effects on gene expression. FGF-10 and its receptors, FGFR1 and FGFR2, have been implicated in breast cancer susceptibility and progression, suggesting that fibroblast growth factor (FGF) signaling may be co-opted by breast cancer cells. We identify a novel pathway downstream of FGFR1 activation, whereby the receptor is cleaved and traffics to the nucleus, where it can regulate specific target genes. We confirm Granzyme B (GrB) as the protease responsible for cleavage and show that blocking GrB activity stopped FGFR1 trafficking to the nucleus and abrogates the promigratory effect of FGF stimulation. We confirm the in vivo relevance of our findings, showing that FGFR1 localized to the nucleus specifically in invading cells in both clinical material and a three-dimensional model of breast cancer. We identify target genes for FGFR1, which exert significant effects on cell migration and may represent an invasive signature. Our experiments identify a novel mechanism by which FGF signaling can regulate cancer cell behavior and provide a novel therapeutic target for treatment of invasive breast cancer.
Collapse
Affiliation(s)
- Athina-Myrto Chioni
- Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, London EC1M 6BQ, England, UK
| | | |
Collapse
|
45
|
Ratzka A, Baron O, Stachowiak MK, Grothe C. Fibroblast growth factor 2 regulates dopaminergic neuron development in vivo. J Neurochem 2012; 122:94-105. [DOI: 10.1111/j.1471-4159.2012.07768.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
46
|
Gibson NJ, Tolbert LP, Oland LA. Activation of glial FGFRs is essential in glial migration, proliferation, and survival and in glia-neuron signaling during olfactory system development. PLoS One 2012; 7:e33828. [PMID: 22493675 PMCID: PMC3320908 DOI: 10.1371/journal.pone.0033828] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2011] [Accepted: 02/22/2012] [Indexed: 11/18/2022] Open
Abstract
Development of the adult olfactory system of the moth Manduca sexta depends on reciprocal interactions between olfactory receptor neuron (ORN) axons growing in from the periphery and centrally-derived glial cells. Early-arriving ORN axons induce a subset of glial cells to proliferate and migrate to form an axon-sorting zone, in which later-arriving ORN axons will change their axonal neighbors and change their direction of outgrowth in order to travel with like axons to their target areas in the olfactory (antennal) lobe. These newly fasciculated axon bundles will terminate in protoglomeruli, the formation of which induces other glial cells to migrate to surround them. Glial cells do not migrate unless ORN axons are present, axons fail to fasciculate and target correctly without sufficient glial cells, and protoglomeruli are not maintained without a glial surround. We have shown previously that Epidermal Growth Factor receptors and the IgCAMs Neuroglian and Fasciclin II play a role in the ORN responses to glial cells. In the present work, we present evidence for the importance of glial Fibroblast Growth Factor receptors in glial migration, proliferation, and survival in this developing pathway. We also report changes in growth patterns of ORN axons and of the dendrites of olfactory (antennal lobe) neurons following blockade of glial FGFR activation that suggest that glial FGFR activation is important in reciprocal communication between neurons and glial cells.
Collapse
Affiliation(s)
- Nicholas J Gibson
- Department of Neuroscience, University of Arizona, Tucson, Arizona, United States of America.
| | | | | |
Collapse
|
47
|
Suzuki A, Harada H, Nakamura H. Nuclear translocation of FGF8 and its implication to induce Sprouty2. Dev Growth Differ 2012; 54:463-73. [PMID: 22404534 DOI: 10.1111/j.1440-169x.2012.01332.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Fibroblast growth factor 8 (FGF8) functions as a local organizing signal for the tectum and cerebellum. FGF8 activates Ras-ERK signaling pathway to induce cerebellar development. We paid attention to the difference in the expression pattern of the molecules that are induced by FGF8 in the mid-hind brain region during normal development and after FGF8 misexpression; some are expressed in the area corresponding to the ERK activation domain but the others are expressed corresponding to the Fgf8 expression domain. Since some of the FGF family members are localized in the nucleus, we wondered if FGF8 could localize in the nuclei and function in the nucleus. We first show that in cultured NIH3T3 cells transfected FGF8b could localize in the nucleus. Transfected FGF8b could also localize in the nucleus of the cells in the chick neural tube. In mouse embryonic neural tube, we detected endogenous FGF8 in the nuclei. Implantation of an FGF8b-soaked bead showed that exogenous FGF8b could be translocated to the nuclei in the isthmus. Furthermore, signal-peptide-deletion mutant of FGF8b mainly localized in the nuclei, and induced Sprouty2 without activating ERK in the mesencephalon. Signal-peptide-deletion mutant of FGF8b could not induce Pax2 expression. Taken together, we concluded that FGF8b could be translocated to the nuclei, and that the nuclear FGF8 could function as transcriptional regulator to induce Sprouty2 in the isthmus.
Collapse
Affiliation(s)
- Ayumu Suzuki
- Department of Molecular Neurobiology, Graduate School of Life Sciences and Institute of Development, Aging and Cancer, Tohoku University, Seiryo-machi 4-1, Aoba-ku, 980-8575 Sendai, Japan
| | | | | |
Collapse
|
48
|
Han W, Lo HW. Landscape of EGFR signaling network in human cancers: biology and therapeutic response in relation to receptor subcellular locations. Cancer Lett 2012; 318:124-34. [PMID: 22261334 DOI: 10.1016/j.canlet.2012.01.011] [Citation(s) in RCA: 185] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2011] [Revised: 01/03/2012] [Accepted: 01/10/2012] [Indexed: 10/14/2022]
Abstract
The epidermal growth factor receptor (EGFR) pathway is one of the most dysregulated molecular pathways in human cancers. Despite its well-established importance in tumor growth, progression and drug-resistant phenotype over the past several decades, targeted therapy designed to circumvent EGFR has yielded only modest clinical success in cancer patients, except those with non-small cell lung cancer (NSCLC) carrying EGFR activation mutations. However, almost all of these NSCLC patients eventually developed resistance to small molecule EGFR kinase inhibitors. These disappointing outcomes are, in part, due to the high complexity and the interactive nature of the EGFR signaling network. More recent compelling evidence further indicates that EGFR functionality can be dependent on its subcellular location. In this regard, EGFR undergoes translocation into different organelles where it elicits distinctly different functions than its best known activity as a plasma membrane-bound receptor tyrosine kinase. EGFR can be shuttled into the cell nucleus and mitochondrion upon ligand binding, radiation, EGFR-targeted therapy and other stimuli. Nuclear EGFR behaves as transcriptional regulator, tyrosine kinase, and mediator of other physiological processes. The role of mitochondrial EGFR remains poorly understood but it appears to regulate apoptosis and autophagy. While studies using patient tumors have shown nuclear EGFR to be an indicator for poor clinical outcomes in cancer patients, the impact of mitochondrial EGFR on tumor behavior and patient prognosis remains to be defined. Most recently, several lines of evidence suggest that mislocated EGFR may regulate tumor response to therapy and that plasma membrane-bound EGFR elicits survival signals independent of its kinase activity. In light of these recent progresses and discoveries, we will outline in this minireview an emerging line of research that uncovers and functionally characterizes several novel modes of EGFR signaling that take center stage in the cell nucleus, mitochondrion and other subcellular compartments. We will also discuss the clinical implications of these findings in the rationale design for therapeutic strategy that overcomes tumor drug resistance.
Collapse
Affiliation(s)
- Woody Han
- Division of Surgical Sciences, Department of Surgery, Duke University School of Medicine, Durham, NC 27710, United States
| | | |
Collapse
|
49
|
Kucinski A, Wersinger S, Stachowiak EK, Radell M, Hesse R, Corso T, Parry M, Bencherif M, Jordan K, Letchworth S, Stachowiak MK. Unilateral 6-OHDA <i>th-fgfr1</i>(<i>tk-</i>) mouse model supports the role of FGFs in Parkinson’s disease and the effects of nicotine and L-DOPA on spontaneous motor impairments. Health (London) 2012. [DOI: 10.4236/health.2012.431176] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
50
|
Lichtenstein MP, Madrigal JLM, Pujol A, Galea E. JNK/ERK/FAK mediate promigratory actions of basic fibroblast growth factor in astrocytes via CCL2 and COX2. Neurosignals 2011; 20:86-102. [PMID: 22189091 DOI: 10.1159/000330805] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2011] [Accepted: 07/07/2011] [Indexed: 12/31/2022] Open
Abstract
While the role of cytokines in causing pro- and anti-inflammatory cascades in the brain and that of chemokines in promoting chemotaxis is well recognized, the immunomodulatory actions of neurotrophins released during brain injury remains largely undetermined. This knowledge gap affects basic fibroblast growth factor (FGF2), which in the brain is mainly produced by astrocytes and characteristically upregulated in reactive astrocytes. The goal of this study was to characterize the inflammatory actions of FGF2 in astrocytes using primary cultures. We report that FGF2 induced the upregulation of monocyte chemoattractant protein (CCL2) and cyclo-oxygenase type 2 (COX2), and the inhibition of lipopolysaccharide-elicited ICAM1 upregulation. The effects of FGF2 were: (i) dependent on gene transcription as revealed by the concomitant regulation of CCL2 or ICAM1 mRNAs; (ii) mediated by the FGF2 receptor type 2; (iii) dependent on ERK, JNK and FAK, and (iv) NF-κB-independent. FGF2 also caused accelerated wound closure dependent on CCL2, COX2, ERK, JNK and FAK in a scratch assay. We conclude that the signaling network triggered by FGF2 in astrocytes converged into accelerating directed motion. It follows that astrocyte migration to injury sites may be a key factor in the repair mechanisms orchestrated by FGF2.
Collapse
|