1
|
Hirashima T, Matsuda M. ERK-mediated curvature feedback regulates branching morphogenesis in lung epithelial tissue. Curr Biol 2024; 34:683-696.e6. [PMID: 38228149 DOI: 10.1016/j.cub.2023.12.049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 11/06/2023] [Accepted: 12/14/2023] [Indexed: 01/18/2024]
Abstract
Intricate branching patterns emerge in internal organs due to the recurrent occurrence of simple deformations in epithelial tissues. During murine lung development, epithelial cells in distal tips of the single tube require fibroblast growth factor (FGF) signals emanating from their surrounding mesenchyme to form repetitive tip bifurcations. However, it remains unknown how the cells employ FGF signaling to convert their behaviors to achieve the recursive branching processes. Here, we show a mechano-chemical regulatory system underlying lung branching morphogenesis, orchestrated by extracellular signal-regulated kinase (ERK) as a downstream driver of FGF signaling. We found that tissue-scale curvature regulated ERK activity in the lung epithelium using two-photon live cell imaging and mechanical perturbations. ERK activation occurs specifically in epithelial tissues exhibiting positive curvature, regardless of whether the change in curvature was attributable to morphogenesis or perturbations. Moreover, ERK activation accelerates actin polymerization preferentially at the apical side of cells, mechanically contributing to the extension of the apical membrane, culminating in a reduction of epithelial tissue curvature. These results indicate the existence of a negative feedback loop between tissue curvature and ERK activity that transcends spatial scales. Our mathematical model confirms that this regulatory mechanism is sufficient to generate the recursive branching processes. Taken together, we propose that ERK orchestrates a curvature feedback loop pivotal to the self-organized patterning of tissues.
Collapse
Affiliation(s)
- Tsuyoshi Hirashima
- Mechanobiology Institute, National University of Singapore, 5A Engineering Drive 1, Singapore 117411, Singapore; Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, 2 Medical Drive MD9, Singapore 117593, Singapore; The Hakubi Center, Kyoto University, Yoshida-honmachi, Kyoto 606-8501, Japan; Graduate School of Biostudies, Kyoto University, Yoshidakone-cho, Kyoto 606-8501, Japan; Japan Science and Technology Agency, PRESTO, 4-1-8 Honchō, Kawaguchi 332-0012, Japan.
| | - Michiyuki Matsuda
- Graduate School of Biostudies, Kyoto University, Yoshidakone-cho, Kyoto 606-8501, Japan; Graduate School of Medicine, Kyoto University, Yoshidakone-cho, Kyoto 606-8501, Japan; Institute for Integrated Cell-Material Sciences, Kyoto University, Yoshida-honmachi, Kyoto 606-8317, Japan
| |
Collapse
|
2
|
Kuczyńska M, Moskot M, Gabig-Cimińska M. Insights into Autophagic Machinery and Lysosomal Function in Cells Involved in the Psoriatic Immune-Mediated Inflammatory Cascade. Arch Immunol Ther Exp (Warsz) 2024; 72:aite-2024-0005. [PMID: 38409665 DOI: 10.2478/aite-2024-0005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 12/08/2023] [Indexed: 02/28/2024]
Abstract
Impaired autophagy, due to the dysfunction of lysosomal organelles, contributes to maladaptive responses by pathways central to the immune system. Deciphering the immune-inflammatory ecosystem is essential, but remains a major challenge in terms of understanding the mechanisms responsible for autoimmune diseases. Accumulating evidence implicates a role that is played by a dysfunctional autophagy-lysosomal pathway (ALP) and an immune niche in psoriasis (Ps), one of the most common chronic skin diseases, characterized by the co-existence of autoimmune and autoinflammatory responses. The dysregulated autophagy associated with the defective lysosomal system is only one aspect of Ps pathogenesis. It probably cannot fully explain the pathomechanism involved in Ps, but it is likely important and should be seriously considered in Ps research. This review provides a recent update on discoveries in the field. Also, it sheds light on how the dysregulation of intracellular pathways, coming from modulated autophagy and endolysosomal trafficking, characteristic of key players of the disease, i.e., skin-resident cells, as well as circulating immune cells, may be responsible for immune impairment and the development of Ps.
Collapse
Affiliation(s)
- Martyna Kuczyńska
- Department of Medical Biology and Genetics, University of Gdańsk, Gdańsk, Poland
| | - Marta Moskot
- Department of Medical Biology and Genetics, University of Gdańsk, Gdańsk, Poland
| | | |
Collapse
|
3
|
Tarvestad-Laise KE, Ceresa BP. Modulating Growth Factor Receptor Signaling to Promote Corneal Epithelial Homeostasis. Cells 2023; 12:2730. [PMID: 38067157 PMCID: PMC10706396 DOI: 10.3390/cells12232730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 11/20/2023] [Accepted: 11/27/2023] [Indexed: 12/18/2023] Open
Abstract
The corneal epithelium is the first anatomical barrier between the environment and the cornea; it is critical for proper light refraction onto the retina and prevents pathogens (e.g., bacteria, viruses) from entering the immune-privileged eye. Trauma to the highly innervated corneal epithelium is extremely painful and if not resolved quickly or properly, can lead to infection and ultimately blindness. The healthy eye produces its own growth factors and is continuously bathed in tear fluid that contains these proteins and other nutrients to maintain the rapid turnover and homeostasis of the ocular surface. In this article, we review the roles of growth factors in corneal epithelial homeostasis and regeneration and some of the limitations to their use therapeutically.
Collapse
Affiliation(s)
- Kate E. Tarvestad-Laise
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY 40202, USA
| | - Brian P. Ceresa
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY 40202, USA
- Department of Ophthalmology and Vision Sciences, University of Louisville, Louisville, KY 40202, USA
| |
Collapse
|
4
|
Tarvestad-Laise K, Ceresa BP. Knockout of c-Cbl/Cbl-b slows c-Met trafficking resulting in enhanced signaling in corneal epithelial cells. J Biol Chem 2023; 299:105233. [PMID: 37690689 PMCID: PMC10622846 DOI: 10.1016/j.jbc.2023.105233] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 08/28/2023] [Accepted: 09/01/2023] [Indexed: 09/12/2023] Open
Abstract
In many cell types, the E3 ubiquitin ligases c-Cbl and Cbl-b induce ligand-dependent ubiquitylation of the hepatocyte growth factor (HGF)-stimulated c-Met receptor and target it for lysosomal degradation. This study determines whether c-Cbl/Cbl-b are negative regulators of c-Met in the corneal epithelium (CE) and if their inhibition can augment c-Met-mediated CE homeostasis. Immortalized human corneal epithelial cells were transfected with Cas9 only (Cas9, control cells) or with Cas9 and c-Cbl/Cbl-b guide RNAs to knockout each gene singularly (-c-Cbl or -Cbl-b cells) or both genes (double KO [DKO] cells) and monitored for their responses to HGF. Cells were assessed for ligand-dependent c-Met ubiquitylation via immunoprecipitation, magnitude, and duration of c-Met receptor signaling via immunoblot and receptor trafficking by immunofluorescence. Single KO cells displayed a decrease in receptor ubiquitylation and an increase in phosphorylation compared to control. DKO cells had no detectable ubiquitylation, had delayed receptor trafficking, and a 2.3-fold increase in c-Met phosphorylation. Based on the observed changes in receptor trafficking and signaling, we examined HGF-dependent in vitro wound healing via live-cell time-lapse microscopy in control and DKO cells. HGF-treated DKO cells healed at approximately twice the rate of untreated cells. From these data, we have generated a model in which c-Cbl/Cbl-b mediate the ubiquitylation of c-Met, which targets the receptor through the endocytic pathway toward lysosomal degradation. In the absence of ubiquitylation, the stimulated receptor stays phosphorylated longer and enhances in vitro wound healing. We propose that c-Cbl and Cbl-b are promising pharmacologic targets for enhancing c-Met-mediated CE re-epithelialization.
Collapse
Affiliation(s)
- Kate Tarvestad-Laise
- Department of Pharmacology and Toxicology (KTL, BPC) and Department of Ophthalmology and Vision Sciences (BPC), University of Louisville, Louisville, Kentucky, USA
| | - Brian P Ceresa
- Department of Pharmacology and Toxicology (KTL, BPC) and Department of Ophthalmology and Vision Sciences (BPC), University of Louisville, Louisville, Kentucky, USA.
| |
Collapse
|
5
|
Watson J, Ferguson HR, Brady RM, Ferguson J, Fullwood P, Mo H, Bexley KH, Knight D, Howell G, Schwartz JM, Smith MP, Francavilla C. Spatially resolved phosphoproteomics reveals fibroblast growth factor receptor recycling-driven regulation of autophagy and survival. Nat Commun 2022; 13:6589. [PMID: 36329028 PMCID: PMC9633600 DOI: 10.1038/s41467-022-34298-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Accepted: 10/19/2022] [Indexed: 11/06/2022] Open
Abstract
Receptor Tyrosine Kinase (RTK) endocytosis-dependent signalling drives cell proliferation and motility during development and adult homeostasis, but is dysregulated in diseases, including cancer. The recruitment of RTK signalling partners during endocytosis, specifically during recycling to the plasma membrane, is still unknown. Focusing on Fibroblast Growth Factor Receptor 2b (FGFR2b) recycling, we reveal FGFR signalling partners proximal to recycling endosomes by developing a Spatially Resolved Phosphoproteomics (SRP) approach based on APEX2-driven biotinylation followed by phosphorylated peptides enrichment. Combining this with traditional phosphoproteomics, bioinformatics, and targeted assays, we uncover that FGFR2b stimulated by its recycling ligand FGF10 activates mTOR-dependent signalling and ULK1 at the recycling endosomes, leading to autophagy suppression and cell survival. This adds to the growing importance of RTK recycling in orchestrating cell fate and suggests a therapeutically targetable vulnerability in ligand-responsive cancer cells. Integrating SRP with other systems biology approaches provides a powerful tool to spatially resolve cellular signalling.
Collapse
Affiliation(s)
- Joanne Watson
- Division of Evolution, Infection and Genomics, School of Biological Science, Faculty of Biology Medicine and Health (FBMH), The University of Manchester, M139PT, Manchester, UK
- Division of Molecular and Cellular Function, School of Biological Science, Faculty of Biology Medicine and Health (FBMH), The University of Manchester, M139PT, Manchester, UK
| | - Harriet R Ferguson
- Division of Molecular and Cellular Function, School of Biological Science, Faculty of Biology Medicine and Health (FBMH), The University of Manchester, M139PT, Manchester, UK
| | - Rosie M Brady
- Division of Cancer Sciences, School of Medical Science, Faculty of Biology Medicine and Health (FBMH), The University of Manchester, Manchester, M20 4GJ, UK
| | - Jennifer Ferguson
- Division of Molecular and Cellular Function, School of Biological Science, Faculty of Biology Medicine and Health (FBMH), The University of Manchester, M139PT, Manchester, UK
| | - Paul Fullwood
- Division of Molecular and Cellular Function, School of Biological Science, Faculty of Biology Medicine and Health (FBMH), The University of Manchester, M139PT, Manchester, UK
| | - Hanyi Mo
- Division of Evolution, Infection and Genomics, School of Biological Science, Faculty of Biology Medicine and Health (FBMH), The University of Manchester, M139PT, Manchester, UK
| | - Katherine H Bexley
- Division of Molecular and Cellular Function, School of Biological Science, Faculty of Biology Medicine and Health (FBMH), The University of Manchester, M139PT, Manchester, UK
| | - David Knight
- Bio-MS Core Research Facility, FBMH, The University of Manchester, M139PT, Manchester, UK
| | - Gareth Howell
- Flow Cytometry Core Research Facility, FBMH, The University of Manchester, M139PT, Manchester, UK
| | - Jean-Marc Schwartz
- Division of Evolution, Infection and Genomics, School of Biological Science, Faculty of Biology Medicine and Health (FBMH), The University of Manchester, M139PT, Manchester, UK
| | - Michael P Smith
- Division of Molecular and Cellular Function, School of Biological Science, Faculty of Biology Medicine and Health (FBMH), The University of Manchester, M139PT, Manchester, UK.
| | - Chiara Francavilla
- Division of Molecular and Cellular Function, School of Biological Science, Faculty of Biology Medicine and Health (FBMH), The University of Manchester, M139PT, Manchester, UK.
- Manchester Breast Centre, Manchester Cancer Research Centre, The University of Manchester, M139PT, Manchester, UK.
| |
Collapse
|
6
|
Francavilla C, O'Brien CS. Fibroblast growth factor receptor signalling dysregulation and targeting in breast cancer. Open Biol 2022; 12:210373. [PMID: 35193394 PMCID: PMC8864352 DOI: 10.1098/rsob.210373] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Accepted: 01/20/2022] [Indexed: 01/07/2023] Open
Abstract
Fibroblast Growth Factor Receptor (FGFR) signalling plays a critical role in breast embryonal development, tissue homeostasis, tumorigenesis and metastasis. FGFR, its numerous FGF ligands and signalling partners are often dysregulated in breast cancer progression and are one of the causes of resistance to treatment in breast cancer. Furthermore, FGFR signalling on epithelial cells is affected by signals from the breast microenvironment, therefore increasing the possibility of breast developmental abnormalities or cancer progression. Increasing our understanding of the multi-layered roles of the complex family of FGFRs, their ligands FGFs and their regulatory partners may offer novel treatment strategies for breast cancer patients, as a single agent or rational co-target, which will be explored in depth in this review.
Collapse
Affiliation(s)
- Chiara Francavilla
- Division of Molecular and Cellular Function, School of Biological Science, Faculty of Biology, Medicine and Health (FBMH), University of Manchester, Manchester M13 9PT, UK
- The Manchester Breast Centre, University of Manchester, Wilmslow Road, Manchester M20 4GJ, UK
| | - Ciara S. O'Brien
- The Christie Hospital NHS Foundation Trust, Wilmslow Road, Manchester M20 2BX, UK
- The Manchester Breast Centre, University of Manchester, Wilmslow Road, Manchester M20 4GJ, UK
| |
Collapse
|
7
|
Bártolo IP, Reis RL, Marques AP, Cerqueira M. Keratinocyte Growth Factor-based Strategies for Wound Re-epithelialization. TISSUE ENGINEERING PART B-REVIEWS 2021; 28:665-676. [PMID: 34238035 DOI: 10.1089/ten.teb.2021.0030] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Wound re-epithelialization is a dynamic process that comprises the formation of new epithelium through an active signaling network between several growth factors and various cell types. The main players are keratinocytes that migrate from the wound edges onto the wound bed, to restore the epidermal barrier. One of the most important molecules involved in the re-epithelialization process is Keratinocyte Growth Factor (KGF), since it is central on promoting both migration and proliferation of keratinocytes. Stromal cells, like dermal fibroblasts, are the main producers of this factor, acting on keratinocytes through paracrine signaling. Multiple therapeutic strategies to delivery KGF have been proposed in order to boost wound healing by targeting re-epithelialization. This has been achieved through a range of different approaches, such as topical application, using controlled release-based methods with different biomaterials (hydrogels, nanoparticles and membranes) and also through gene therapy techniques. Among these strategies, KGF delivery via biomaterials and genetic-based strategies show great effectiveness in sustained KGF levels at the wound site, leading to efficient wound closure. Under this scope, this review aims at highlighting the importance of KGF as one of the key molecules on wound re-epithelialization, as well as to provide a critical overview of the different potential therapeutic strategies exploited so far.
Collapse
Affiliation(s)
- Inês P Bártolo
- 3B's Research Group, 226382, Barco, Portugal.,Laboratorio Associado ICVS 3B's, 511313, Guimaraes, Portugal;
| | - Rui L Reis
- 3B's Research Group, 226382, Guimaraes, Portugal.,Laboratorio Associado ICVS 3B's, 511313, Braga/Guimaraes, Portugal;
| | - Alexandra P Marques
- 3B's Research Group, 226382, Guimaraes, Portugal.,Laboratorio Associado ICVS 3B's, 511313, Braga/Guimaraes, Portugal;
| | - Mariana Cerqueira
- 3B's Research Group, 226382, Guimaraes, Portugal.,Laboratorio Associado ICVS 3B's, 511313, Braga/Guimaraes, Portugal;
| |
Collapse
|
8
|
Seitz T, Hellerbrand C. Role of fibroblast growth factor signalling in hepatic fibrosis. Liver Int 2021; 41:1201-1215. [PMID: 33655624 DOI: 10.1111/liv.14863] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 02/22/2021] [Accepted: 02/25/2021] [Indexed: 12/11/2022]
Abstract
Fibrotic remodelling is a highly conserved protective response to tissue injury and it is essential for the maintenance of structural and functional tissue integrity. Also hepatic fibrosis can be considered as a wound-healing response to liver injury, reflecting a balance between liver repair and scar formation. In contrast, pathological fibrosis corresponds to impaired wound healing. Usually, the liver regenerates after acute injury. However, if the damaging mechanisms persist, the liver reacts with progressive and uncontrolled accumulation of extracellular matrix proteins. Eventually, excessive fibrosis can lead to cirrhosis and hepatic failure. Furthermore, cirrhosis is the major risk factor for the development of hepatocellular cancer (HCC). Therefore, hepatic fibrosis is the most critical pathological factor that determines the morbidity and mortality of patients with chronic liver disease. Still, no effective anti-fibrogenic therapies exist, despite the very high medical need. The regulation of fibroblast growth factor (FGF) signalling is a prerequisite for adequate wound healing, repair and homeostasis in various tissues and organs. The FGF family comprises 22 proteins that can be classified into paracrine, intracrine and endocrine factors. Most FGFs signal through transmembrane tyrosine kinase FGF receptors (FGFRs). Although FGFRs are promising targets for the treatment of HCC, the expression and function of FGFR-ligands in hepatic fibrosis is still poorly understood. This review summarizes the latest advances in our understanding of FGF signalling in hepatic fibrosis. Furthermore, the potential of FGFs as targets for the treatment of hepatic fibrosis and remaining challenges for the field are discussed.
Collapse
Affiliation(s)
- Tatjana Seitz
- Institute of Biochemistry, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Claus Hellerbrand
- Institute of Biochemistry, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
9
|
Szybowska P, Kostas M, Wesche J, Haugsten EM, Wiedlocha A. Negative Regulation of FGFR (Fibroblast Growth Factor Receptor) Signaling. Cells 2021; 10:cells10061342. [PMID: 34071546 PMCID: PMC8226934 DOI: 10.3390/cells10061342] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 05/25/2021] [Accepted: 05/25/2021] [Indexed: 02/06/2023] Open
Abstract
FGFR (fibroblast growth factor receptor) signaling controls fundamental processes in embryonic, fetal and adult human life. The magnitude, duration, and location of FGFR signaling must be strictly controlled in order to induce the correct biological response. Uncontrolled receptor signaling has been shown to lead to a variety of diseases, such as skeletal disorders and cancer. Here we review the numerous cellular mechanisms that regulate and turn off FGFR signaling, once the receptor is activated. These mechanisms include endocytosis and endocytic sorting, phosphatase activity, negative regulatory proteins and negative feedback phosphorylation events. The mechanisms act together simultaneously or sequentially, controlling the same or different steps in FGFR signaling. Although more work is needed to fully understand the regulation of FGFR signaling, it is clear that the cells in our body have evolved an extensive repertoire of mechanisms that together keep FGFR signaling tightly controlled and prevent excess FGFR signaling.
Collapse
Affiliation(s)
- Patrycja Szybowska
- Department of Tumor Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Montebello, 0379 Oslo, Norway; (P.S.); (M.K.); (J.W.)
- Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Montebello, 0379 Oslo, Norway
| | - Michal Kostas
- Department of Tumor Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Montebello, 0379 Oslo, Norway; (P.S.); (M.K.); (J.W.)
- Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Montebello, 0379 Oslo, Norway
| | - Jørgen Wesche
- Department of Tumor Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Montebello, 0379 Oslo, Norway; (P.S.); (M.K.); (J.W.)
- Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Montebello, 0379 Oslo, Norway
| | - Ellen Margrethe Haugsten
- Department of Tumor Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Montebello, 0379 Oslo, Norway; (P.S.); (M.K.); (J.W.)
- Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Montebello, 0379 Oslo, Norway
- Correspondence: (E.M.H.); (A.W.); Tel.: +47-2278-1785 (E.M.H.); +47-2278-1930 (A.W.)
| | - Antoni Wiedlocha
- Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Montebello, 0379 Oslo, Norway
- Department of Molecular Cell Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Montebello, 0379 Oslo, Norway
- Military Institute of Hygiene and Epidemiology, 01-163 Warsaw, Poland
- Correspondence: (E.M.H.); (A.W.); Tel.: +47-2278-1785 (E.M.H.); +47-2278-1930 (A.W.)
| |
Collapse
|
10
|
Ferguson HR, Smith MP, Francavilla C. Fibroblast Growth Factor Receptors (FGFRs) and Noncanonical Partners in Cancer Signaling. Cells 2021; 10:1201. [PMID: 34068954 PMCID: PMC8156822 DOI: 10.3390/cells10051201] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 05/06/2021] [Accepted: 05/09/2021] [Indexed: 02/07/2023] Open
Abstract
Increasing evidence indicates that success of targeted therapies in the treatment of cancer is context-dependent and is influenced by a complex crosstalk between signaling pathways and between cell types in the tumor. The Fibroblast Growth Factor (FGF)/FGF receptor (FGFR) signaling axis highlights the importance of such context-dependent signaling in cancer. Aberrant FGFR signaling has been characterized in almost all cancer types, most commonly non-small cell lung cancer (NSCLC), breast cancer, glioblastoma, prostate cancer and gastrointestinal cancer. This occurs primarily through amplification and over-expression of FGFR1 and FGFR2 resulting in ligand-independent activation. Mutations and translocations of FGFR1-4 are also identified in cancer. Canonical FGF-FGFR signaling is tightly regulated by ligand-receptor combinations as well as direct interactions with the FGFR coreceptors heparan sulfate proteoglycans (HSPGs) and Klotho. Noncanonical FGFR signaling partners have been implicated in differential regulation of FGFR signaling. FGFR directly interacts with cell adhesion molecules (CAMs) and extracellular matrix (ECM) proteins, contributing to invasive and migratory properties of cancer cells, whereas interactions with other receptor tyrosine kinases (RTKs) regulate angiogenic, resistance to therapy, and metastatic potential of cancer cells. The diversity in FGFR signaling partners supports a role for FGFR signaling in cancer, independent of genetic aberration.
Collapse
Affiliation(s)
- Harriet R. Ferguson
- Division of Molecular and Cellular Function, School of Biological Science, Faculty of Biology Medicine and Health (FBMH), The University of Manchester, Manchester M13 9PT, UK;
| | - Michael P. Smith
- Division of Molecular and Cellular Function, School of Biological Science, Faculty of Biology Medicine and Health (FBMH), The University of Manchester, Manchester M13 9PT, UK;
| | - Chiara Francavilla
- Division of Molecular and Cellular Function, School of Biological Science, Faculty of Biology Medicine and Health (FBMH), The University of Manchester, Manchester M13 9PT, UK;
- Manchester Breast Centre, Manchester Cancer Research Centre, The University of Manchester, Manchester M20 4GJ, UK
| |
Collapse
|
11
|
Bharti S, Vadlamudi HC. A strategic review on the involvement of receptors, transcription factors and hormones in acne pathogenesis. J Recept Signal Transduct Res 2020; 41:105-116. [PMID: 32787477 DOI: 10.1080/10799893.2020.1805626] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Acne vulgaris is a very common pilosebaceous inflammatory disease occurring primarily on the face and also rare on the upper arms, trunk, and back, which is caused by Propionibacterium, Staphylococcus, Corynebacterium, and other species. Pathophysiology of acne comprises of irregular keratinocyte proliferation, differentiation, increased sebum output, bacterial antigens and cytokines induced inflammatory response. Treatment of acne requires proper knowledge on the pathophysiology then only the clinician can come out with a proper therapeutic dosage regimen. Understanding the pathophysiology not only includes the mechanism but also involvement of receptors. Thus, this review is framed in such a way that the authors have focused on the disease acne vulgaris, pathophysiology, transcription factors viz. the Forkhead Box O1 (FoxO1) Transcription Factor, hormones like androgens and receptors such as Histamine receptors, Retinoic receptor, Fibroblast growth factor receptors, Toll like receptor, Androgen receptor, Liver X-receptor, Melanocortin receptor, Peroxisome proliferator-activated receptor and epidermal growth factor receptors involvement in the progression of acne vulgaris.
Collapse
Affiliation(s)
- Sneha Bharti
- Department of Pharmaceutics, Acharya & BM Reddy College of Pharmacy, Bangalore, India
| | | |
Collapse
|
12
|
Kang D, Jung SH, Lee GH, Lee S, Park HJ, Ko YG, Kim YN, Lee JS. Sulfated syndecan 1 is critical to preventing cellular senescence by modulating fibroblast growth factor receptor endocytosis. FASEB J 2020; 34:10316-10328. [PMID: 32530114 DOI: 10.1096/fj.201902714r] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 05/06/2020] [Accepted: 05/20/2020] [Indexed: 01/10/2023]
Abstract
Cellular senescence can be triggered by various intrinsic and extrinsic stimuli. We previously reported that silencing of 3'-phosphoadenosine 5'-phosphosulfate synthetase 2 (PAPSS2) induces cellular senescence through augmented fibroblast growth factor receptor 1 (FGFR1) signaling. However, the exact molecular mechanism connecting heparan sulfation and cellular senescence remains unclear. Here, we investigated the potential involvement of heparan sulfate proteoglycans (HSPGs) in augmented FGFR1 signaling and cellular senescence. Depletion of several types of HSPGs revealed that cells depleted of syndecan 1 (SDC1) exhibited typical senescence phenotypes, and those depleted of PAPSS2-, SDC1-, or heparan sulfate 2-O sulfotransferase 1 (HS2ST1) showed decreased FGFR1 internalization along with hyperresponsiveness to and prolonged activation of fibroblast growth factor 2 (FGF2)-stimulated FGFR1- v-akt murine thymoma viral oncogene homolog (AKT) signaling. Clathrin- and caveolin-mediated FGFR1 endocytosis contributed to cellular senescence through the FGFR1-AKT-p53-p21 signaling pathway. Dynasore treatment triggered senescence phenotypes, augmented FGFR1-AKT-p53-p21 signaling, and decreased SDC1 expression. Finally, the replicatively and prematurely senescent cells were characterized by decreases of SDC1 expression and FGFR1 internalization, and an increase in FGFR1-AKT-p53-p21 signaling. Together, our results demonstrate that properly sulfated SDC1 plays a critical role in preventing cellular senescence through the regulation of FGFR1 endocytosis.
Collapse
Affiliation(s)
- Donghee Kang
- Department of Molecular Medicine, Inha University College of Medicine, Incheon, Korea.,Medical Research Center, Inha University College of Medicine, Incheon, Korea
| | - Seung Hee Jung
- Department of Molecular Medicine, Inha University College of Medicine, Incheon, Korea.,Medical Research Center, Inha University College of Medicine, Incheon, Korea
| | - Gun-Hee Lee
- Department of Molecular Medicine, Inha University College of Medicine, Incheon, Korea.,Medical Research Center, Inha University College of Medicine, Incheon, Korea
| | - Seongju Lee
- Medical Research Center, Inha University College of Medicine, Incheon, Korea.,Department of Anatomy, Inha University College of Medicine, Incheon, Korea
| | - Heon Joo Park
- Medical Research Center, Inha University College of Medicine, Incheon, Korea.,Department of Microbiology, Inha University College of Medicine, Incheon, Korea
| | - Young-Gyu Ko
- Division of Life Sciences, Korea University, Seoul, Korea
| | - Yong-Nyun Kim
- Division of Translational Science, National Cancer Center, Goyang, Korea
| | - Jae-Seon Lee
- Department of Molecular Medicine, Inha University College of Medicine, Incheon, Korea.,Medical Research Center, Inha University College of Medicine, Incheon, Korea
| |
Collapse
|
13
|
Gulberti S, Mao X, Bui C, Fournel-Gigleux S. The role of heparan sulfate maturation in cancer: A focus on the 3O-sulfation and the enigmatic 3O-sulfotransferases (HS3STs). Semin Cancer Biol 2020; 62:68-85. [DOI: 10.1016/j.semcancer.2019.10.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Revised: 10/10/2019] [Accepted: 10/11/2019] [Indexed: 01/05/2023]
|
14
|
Liu L, Lu J, Li X, Wu A, Wu Q, Zhao M, Tang N, Song H. The LIS1/NDE1 Complex Is Essential for FGF Signaling by Regulating FGF Receptor Intracellular Trafficking. Cell Rep 2019; 22:3277-3291. [PMID: 29562183 DOI: 10.1016/j.celrep.2018.02.077] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Revised: 01/23/2018] [Accepted: 02/21/2018] [Indexed: 11/27/2022] Open
Abstract
Intracellular transport of membranous organelles and protein complexes to various destinations is fundamental to signaling transduction and cellular function. The cytoplasmic dynein motor and its regulatory proteins LIS1 and NDE1 are required for transporting a variety of cellular cargos along the microtubule network. In this study, we show that deletion of Lis1 in developing lung endoderm and limb mesenchymal cells causes agenesis of the lungs and limbs. In both mutants, there is increased cell death and decreased fibroblast growth factor (FGF) signaling activity. Mechanistically, LIS1 and its interacting protein NDE1/NDEL1 are associated with FGF receptor-containing vesicles and regulate FGF receptor intracellular trafficking and degradation. Notably, FGF signaling promotes NDE1 tyrosine phosphorylation, which leads to dissociation of LIS1/NDE1 complex. Thus, our studies identify the LIS1/NDE1 complex as an important FGF signaling regulator and provide insights into the bidirectional regulation of cell signaling and transport machinery for endocytosis.
Collapse
Affiliation(s)
- Liansheng Liu
- Life Sciences Institute and Innovation Center for Cell Signaling Network, Zhejiang University, Hangzhou 310058, China
| | - Jinqiu Lu
- Life Sciences Institute and Innovation Center for Cell Signaling Network, Zhejiang University, Hangzhou 310058, China
| | - Xiaoling Li
- Life Sciences Institute and Innovation Center for Cell Signaling Network, Zhejiang University, Hangzhou 310058, China
| | - Ailing Wu
- Life Sciences Institute and Innovation Center for Cell Signaling Network, Zhejiang University, Hangzhou 310058, China
| | - Qingzhe Wu
- Life Sciences Institute and Innovation Center for Cell Signaling Network, Zhejiang University, Hangzhou 310058, China
| | - Mujun Zhao
- State Key Laboratory of Molecular Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Nan Tang
- National Institute of Biological Sciences, Beijing 102206, China
| | - Hai Song
- Life Sciences Institute and Innovation Center for Cell Signaling Network, Zhejiang University, Hangzhou 310058, China.
| |
Collapse
|
15
|
Lamberti D, Cristinziano G, Porru M, Leonetti C, Egan JB, Shi CX, Buglioni S, Amoreo CA, Castellani L, Borad MJ, Alemà S, Anastasi S, Segatto O. HSP90 Inhibition Drives Degradation of FGFR2 Fusion Proteins: Implications for Treatment of Cholangiocarcinoma. Hepatology 2019; 69:131-142. [PMID: 30067876 DOI: 10.1002/hep.30127] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Accepted: 05/31/2018] [Indexed: 12/14/2022]
Abstract
About 15% of intrahepatic cholangiocarcinomas (ICCs) express constitutively active fibroblast growth factor receptor 2 (FGFR2) fusion proteins (FFs) generated by chromosomal translocations. FFs have been nominated as oncogenic drivers because administration of FGFR tyrosine kinase inhibitors (F-TKIs) can elicit meaningful objective clinical responses in patients carrying FF-positive ICC. Thus, optimization of FF targeting is a pressing clinical need. Herein, we report that three different FFs, previously isolated from ICC samples, are heat shock protein 90 (HSP90) clients and undergo rapid degradation upon HSP90 pharmacological blockade by the clinically advanced HSP90 inhibitor ganetespib. Combining catalytic suppression by the F-TKI BGJ398 with HSP90 blockade by ganetespib suppressed FGFR2-TACC3 (transforming acidic coiled-coil containing protein 3) signaling in cultured cells more effectively than either BGJ398 or ganetespib in isolation. The BGJ398 + ganetespib combo was also superior to single agents when tested in mice carrying subcutaneous tumors generated by transplantation of FGFR2-TACC3 NIH3T3 transformants. Of note, FF mutants known to enforce clinical resistance to BGJ398 in ICC patients retained full sensitivity to ganetespib in cultured cells. Conclusion: Our data provide a proof of principle that upfront treatment with the BGJ398 + ganetespib combo improves therapeutic targeting of FGFR2 fusions in an experimental setting, which may be relevant to precision medicine approaches to FF-driven ICC.
Collapse
Affiliation(s)
- Dante Lamberti
- Unit of Oncogenomics and Epigenetics, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Giulia Cristinziano
- Unit of Oncogenomics and Epigenetics, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Manuela Porru
- Animal Facility (SAFU), IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Carlo Leonetti
- Animal Facility (SAFU), IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Jan B Egan
- Division of Hematology and Oncology, Mayo Clinic, Scottsdale, Arizona
| | - Chang-Xin Shi
- Division of Hematology and Oncology, Mayo Clinic, Scottsdale, Arizona
| | - Simonetta Buglioni
- Department of Pathology, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Carla A Amoreo
- Department of Pathology, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Loriana Castellani
- Dipartimento di Scienze Umane, Sociali e della Salute, Università di Cassino, Cassino, Italy.,Institute of Cell Biology and Neurobiology, National Research Council (CNR), Monterotondo, Italy
| | - Mitesh J Borad
- Division of Hematology and Oncology, Mayo Clinic, Scottsdale, Arizona
| | - Stefano Alemà
- Institute of Cell Biology and Neurobiology, National Research Council (CNR), Monterotondo, Italy
| | - Sergio Anastasi
- Unit of Oncogenomics and Epigenetics, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Oreste Segatto
- Unit of Oncogenomics and Epigenetics, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| |
Collapse
|
16
|
Watson J, Francavilla C. Regulation of FGF10 Signaling in Development and Disease. Front Genet 2018; 9:500. [PMID: 30405705 PMCID: PMC6205963 DOI: 10.3389/fgene.2018.00500] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Accepted: 10/05/2018] [Indexed: 12/12/2022] Open
Abstract
Fibroblast Growth Factor 10 (FGF10) is a multifunctional mesenchymal-epithelial signaling growth factor, which is essential for multi-organ development and tissue homeostasis in adults. Furthermore, FGF10 deregulation has been associated with human genetic disorders and certain forms of cancer. Upon binding to FGF receptors with heparan sulfate as co-factor, FGF10 activates several intracellular signaling cascades, resulting in cell proliferation, differentiation, and invasion. FGF10 activity is modulated not only by heparan sulfate proteoglycans in the extracellular matrix, but also by hormones and other soluble factors. Despite more than 20 years of research on FGF10 functions, context-dependent regulation of FGF10 signaling specificity remains poorly understood. Emerging modes of FGF10 signaling regulation will be described, focusing on the role of FGF10 trafficking and sub-cellular localization, heparan sulfate proteoglycans, and miRNAs. Systems biology approaches based on quantitative proteomics will be considered for globally investigating FGF10 signaling specificity. Finally, current gaps in our understanding of FGF10 functions, such as the relative contribution of receptor isoforms to signaling activation, will be discussed in the context of genetic disorders and tumorigenesis.
Collapse
Affiliation(s)
- Joanne Watson
- Division of Molecular and Cellular Function, School of Biological Sciences, Faculty of Biology Medicine and Health, The University of Manchester, Manchester, United Kingdom
- Division of Evolution and Genomic Sciences, School of Biological Sciences, Faculty of Biology Medicine and Health, The University of Manchester, Manchester, United Kingdom
| | - Chiara Francavilla
- Division of Molecular and Cellular Function, School of Biological Sciences, Faculty of Biology Medicine and Health, The University of Manchester, Manchester, United Kingdom
| |
Collapse
|
17
|
Zinkle A, Mohammadi M. A threshold model for receptor tyrosine kinase signaling specificity and cell fate determination. F1000Res 2018; 7:F1000 Faculty Rev-872. [PMID: 29983915 PMCID: PMC6013765 DOI: 10.12688/f1000research.14143.1] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/18/2018] [Indexed: 11/20/2022] Open
Abstract
Upon ligand engagement, the single-pass transmembrane receptor tyrosine kinases (RTKs) dimerize to transmit qualitatively and quantitatively different intracellular signals that alter the transcriptional landscape and thereby determine the cellular response. The molecular mechanisms underlying these fundamental events are not well understood. Considering recent insights into the structural biology of fibroblast growth factor signaling, we propose a threshold model for RTK signaling specificity in which quantitative differences in the strength/longevity of ligand-induced receptor dimers on the cell surface lead to quantitative differences in the phosphorylation of activation loop (A-loop) tyrosines as well as qualitative differences in the phosphorylation of tyrosines mediating substrate recruitment. In this model, quantitative differences on A-loop tyrosine phosphorylation result in gradations in kinase activation, leading to the generation of intracellular signals of varying amplitude/duration. In contrast, qualitative differences in the pattern of tyrosine phosphorylation on the receptor result in the recruitment/activation of distinct substrates/intracellular pathways. Commensurate with both the dynamics of the intracellular signal and the types of intracellular pathways activated, unique transcriptional signatures are established. Our model provides a framework for engineering clinically useful ligands that can tune receptor dimerization stability so as to bias the cellular transcriptome to achieve a desired cellular output.
Collapse
Affiliation(s)
- Allen Zinkle
- Department of Biochemistry & Molecular Pharmacology, New York University School of Medicine, New York, NY, USA
| | - Moosa Mohammadi
- Department of Biochemistry & Molecular Pharmacology, New York University School of Medicine, New York, NY, USA
| |
Collapse
|
18
|
Neuropilin 1 Mediates Keratinocyte Growth Factor Signaling in Adipose-Derived Stem Cells: Potential Involvement in Adipogenesis. Stem Cells Int 2018. [PMID: 29535768 PMCID: PMC5845512 DOI: 10.1155/2018/1075156] [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] [Indexed: 12/16/2022] Open
Abstract
Adipogenesis is regulated by a complex network of molecules, including fibroblast growth factors. Keratinocyte growth factor (KGF) has been previously reported to promote proliferation on rat preadipocytes, although the expression of its specific receptor, FGFR2-IIIb/KGFR, is not actually detected in mesenchymal cells. Here, we demonstrate that human adipose-derived stem cells (ASCs) show increased expression of KGF during adipogenic differentiation, especially in the early steps. Moreover, KGF is able to induce transient activation of ERK and p38 MAPK pathways in these cells. Furthermore, KGF promotes ASC differentiation and supports the activation of differentiation pathways, such as those of PI3K/Akt and the retinoblastoma protein (Rb). Notably, we observed only a low amount of FGFR2-IIIb in ASCs, which seems not to be responsible for KGF activity. Here, we demonstrate for the first time that Neuropilin 1 (NRP1), a transmembrane glycoprotein expressed in ASCs acting as a coreceptor for some growth factors, is responsible for KGF-dependent pathway activation in these cells. Our study contributes to clarify the molecular bases of human adipogenesis, demonstrating a role of KGF in the early steps of this process, and points out a role of NRP1 as a previously unknown mediator of KGF action in ASCs.
Collapse
|
19
|
Nakamura R, Tani A, Yoshie S, Ikeda M, Wada I, Hazama A, Nomoto Y, Tada Y, Nakamura T, Omori K. Heparin cross-linked collagen sponge scaffolds improve functional regeneration of rat tracheal epithelium. J Tissue Eng Regen Med 2017; 11:3027-3037. [PMID: 28101973 DOI: 10.1002/term.2204] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Revised: 03/09/2016] [Accepted: 03/27/2016] [Indexed: 11/12/2022]
Abstract
Tracheal epithelial cells maintain airway homeostasis by mediating mucociliary clearance. Following tracheal reconstruction, timely epithelial regeneration is required to prevent respiratory compromise and infectious diseases. To achieve rapid tracheal epithelial regeneration, a heparin cross-linked collagen sponge containing fibroblast growth factor-2 (FGF-2) was prepared as a graft for tracheal reconstruction. The heparin cross-linked sponge exhibited a high FGF-2 retaining capacity, and tracheal epithelial and mesenchymal cells cultured in this sponge containing FGF-2 showed high proliferative capacities. Subsequently, heparin-free collagen sponge scaffolds (C/F scaffold) and collagen sponge scaffolds cross-linked with 10 μg/ml heparin retained FGF-2 (C/H10/F scaffold), and were transplanted into rats with tracheal defects. Invasion of both epithelial and non-epithelial cells was greater in rats treated with the C/H10/F scaffold at 1 week post-transplantation than in rats treated with the C/F scaffold. Moreover, at 2 weeks after transplantation, improved cilia formation was observed in the C/H10/F scaffold group, with higher motility and more potent posterior-anterior flow generation than in the C/F scaffold group. These results suggest that heparin improves functional regeneration of tracheal epithelium. Copyright © 2017 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Ryosuke Nakamura
- Department of Otolaryngology, School of Medicine, Fukushima Medical University, Fukushima, City, Japan
| | - Akiko Tani
- Department of Otolaryngology, School of Medicine, Fukushima Medical University, Fukushima, City, Japan
| | - Susumu Yoshie
- Department of Otolaryngology, School of Medicine, Fukushima Medical University, Fukushima, City, Japan
| | - Masakazu Ikeda
- Department of Otolaryngology, School of Medicine, Fukushima Medical University, Fukushima, City, Japan
| | - Ikuo Wada
- Department of Cell Science, Institute of Biomedical Sciences, School of Medicine, Fukushima Medical University, Japan
| | - Akihiro Hazama
- Department of Cellular and Integrative Physiology, School of Medicine, Fukushima Medical University, Japan
| | - Yukio Nomoto
- Department of Otolaryngology, School of Medicine, Fukushima Medical University, Fukushima, City, Japan
| | - Yasuhiro Tada
- Department of Otolaryngology, School of Medicine, Fukushima Medical University, Fukushima, City, Japan
| | - Tatsuo Nakamura
- Department of Bioartificial Organs, Institute for Frontier Medical Sciences, Kyoto University, Japan
| | - Koichi Omori
- Department of Otolaryngology, School of Medicine, Fukushima Medical University, Fukushima, City, Japan
| |
Collapse
|
20
|
Al-Hammadi A, Al-Ismaily A, Al-Ali S, Ramadurai R, Jain R, McKinley-Grant L, Mughal TI. Topical, Biological and Clinical Challenges in the Management of Patients with Acne Vulgaris. Sultan Qaboos Univ Med J 2016; 16:e152-60. [PMID: 27226905 DOI: 10.18295/squmj.2016.16.02.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2014] [Revised: 07/02/2015] [Accepted: 01/18/2016] [Indexed: 11/16/2022] Open
Abstract
Acne vulgaris is one of the most common chronic inflammatory skin disorders among adolescents and young adults. It is associated with substantial morbidity and, rarely, with mortality. The exact worldwide incidence and prevalence are currently unknown. Current challenges involve improving understanding of the underlying pathophysiology of acne vulgaris and developing a practical treatment consensus. Expert panel discussions were held in 2013 and 2014 among a group of scientists and clinicians from the Omani and United Arab Emirate Dermatology Societies to ascertain the current optimal management of acne vulgaris, identify clinically relevant end-points and construct suitable methodology for future clinical trial designs. This article reviews the discussions of these sessions and recent literature on this topic.
Collapse
Affiliation(s)
- Anwar Al-Hammadi
- Department of Dermatology, Rashid Hospital, Dubai, United Arab Emirates
| | | | - Sameer Al-Ali
- Department of Dermatology, Abu Dhabi Dermatology Clinic, Abu Dhabi, United Arab Emirates
| | - Rajesh Ramadurai
- Department of Clinical & Medical Affairs, Glenmark Pharmaceuticals Private Ltd., Mumbai, India
| | - Rishi Jain
- Department of Clinical & Medical Affairs, Glenmark Pharmaceuticals Private Ltd., Mumbai, India
| | - Lynn McKinley-Grant
- Department of Dermatology, George Washington University Hospital, Washington D.C., USA
| | - Tariq I Mughal
- Department of Hematology & Oncology, Tufts University Medical Center, Boston, Massachusetts, USA
| |
Collapse
|
21
|
Abstract
Fibroblast growth factors (FGFs) have been shown to alter growth and differentiation of reproductive tissues in a variety of species. Within the female reproductive tract, the effects of FGFs have been focused on the ovary, and the most studied one is FGF2, which stimulates granulosa cell proliferation and decreases differentiation (decreased steroidogenesis). Other FGFs have also been implicated in ovarian function, and this review summarizes the effects of members of two subfamilies on ovarian function; the FGF7 subfamily that also contains FGF10, and the FGF8 subfamily that also contains FGF18. There are data to suggest that FGF8 and FGF18 have distinct actions on granulosa cells, despite their apparent similar receptor binding properties. Studies of non-reproductive developmental biology also indicate that FGF8 is distinct from FGF18, and that FGF7 is also distinct from FGF10 despite similar receptor binding properties. In this review, the potential mechanisms of differential action of FGF7/FGF10 and FGF8/FGF18 during organogenesis will be reviewed and placed in the context of follicle development. A model is proposed in which FGF8 and FGF18 differentially activate receptors depending on the properties of the extracellular matrix in the follicle.
Collapse
Affiliation(s)
- Christopher A Price
- Faculty of Veterinary MedicineCentre de recherche en reproduction animale, University of Montreal, 3200 rue Sicotte, St-Hyacinthe, Quebec, Canada J2S 7C6
| |
Collapse
|
22
|
Xu J, Liu H, Lan Y, Aronow BJ, Kalinichenko VV, Jiang R. A Shh-Foxf-Fgf18-Shh Molecular Circuit Regulating Palate Development. PLoS Genet 2016; 12:e1005769. [PMID: 26745863 PMCID: PMC4712829 DOI: 10.1371/journal.pgen.1005769] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Accepted: 12/03/2015] [Indexed: 12/20/2022] Open
Abstract
Cleft palate is among the most common birth defects in humans. Previous studies have shown that Shh signaling plays critical roles in palate development and regulates expression of several members of the forkhead-box (Fox) family transcription factors, including Foxf1 and Foxf2, in the facial primordia. Although cleft palate has been reported in mice deficient in Foxf2, whether Foxf2 plays an intrinsic role in and how Foxf2 regulates palate development remain to be elucidated. Using Cre/loxP-mediated tissue-specific gene inactivation in mice, we show that Foxf2 is required in the neural crest-derived palatal mesenchyme for normal palatogenesis. We found that Foxf2 mutant embryos exhibit altered patterns of expression of Shh, Ptch1, and Shox2 in the developing palatal shelves. Through RNA-seq analysis, we identified over 150 genes whose expression was significantly up- or down-regulated in the palatal mesenchyme in Foxf2-/- mutant embryos in comparison with control littermates. Whole mount in situ hybridization analysis revealed that the Foxf2 mutant embryos exhibit strikingly corresponding patterns of ectopic Fgf18 expression in the palatal mesenchyme and concomitant loss of Shh expression in the palatal epithelium in specific subdomains of the palatal shelves that correlate with where Foxf2, but not Foxf1, is expressed during normal palatogenesis. Furthermore, tissue specific inactivation of both Foxf1 and Foxf2 in the early neural crest cells resulted in ectopic activation of Fgf18 expression throughout the palatal mesenchyme and dramatic loss of Shh expression throughout the palatal epithelium. Addition of exogenous Fgf18 protein to cultured palatal explants inhibited Shh expression in the palatal epithelium. Together, these data reveal a novel Shh-Foxf-Fgf18-Shh circuit in the palate development molecular network, in which Foxf1 and Foxf2 regulate palatal shelf growth downstream of Shh signaling, at least in part, by repressing Fgf18 expression in the palatal mesenchyme to ensure maintenance of Shh expression in the palatal epithelium. Cleft lip and/or cleft palate (CL/P) are among the most common birth defects in humans, occurring at a frequency of about 1 in 500–2500 live births. The etiology and pathogenesis of CL/P are complex and poorly understood. Generation and analysis of mice carrying targeted null and conditional mutations in many genes have revealed that functional disruption of each of more than 100 genes could cause cleft palate. However, how these genes work together to regulate palate development is not well understood. In this study, we identify a novel molecular circuit consisting of two critical molecular pathways, the fibroblast growth factor (FGF) and Sonic hedgehog (SHH) signaling pathways, and the Forkhead family transcription factors Foxf1 and Foxf2, mediating reciprocal epithelial-mesenchymal signaling interactions that control palatogenesis. As mutations affecting each of multiple components of both the FGF and SHH signaling pathways have been associated with CL/P in humans, our results provide significant new insight into the mechanisms regulating palatogenesis and cleft palate pathogenesis.
Collapse
Affiliation(s)
- Jingyue Xu
- Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, United States of America
| | - Han Liu
- Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, United States of America
| | - Yu Lan
- Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, United States of America
- Division of Plastic Surgery, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, United States of America
| | - Bruce J. Aronow
- Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, United States of America
- Division of Biomedical Informatics, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, United States of America
| | - Vladimir V. Kalinichenko
- Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, United States of America
- Division of Pulmonary Biology, Perinatal Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, United States of America
| | - Rulang Jiang
- Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, United States of America
- Division of Plastic Surgery, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, United States of America
- * E-mail:
| |
Collapse
|
23
|
Nakamura R, Nakamura F, Fukunaga S. Perlecan Diversely Regulates the Migration and Proliferation of Distinct Cell Types in vitro. Cells Tissues Organs 2015; 200:374-93. [PMID: 26562025 DOI: 10.1159/000440950] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/08/2015] [Indexed: 11/19/2022] Open
Abstract
Perlecan is a multifunctional component of the extracellular matrix. It shows different effects on distinct cell types, and therefore it is thought to show potential for therapies targeting multiple cell types. However, the full range of multifunctionality of perlecan remains to be elucidated. We cultured various cell types, which were derived from epithelial/endothelial, connective and muscle tissues, in the presence of either antiserum against perlecan or exogenous perlecan, and examined the effects of perlecan on cell migration and proliferation. Cell migration was determined using a scratch assay. Blocking of perlecan by anti-perlecan antiserum inhibited the migration of vascular endothelial cells (VECs) and bone marrow-derived mesenchymal stem cells, and exogenous perlecan added to the culture medium promoted the migration of these cell types. The migration of other cell types was inhibited or was not promoted by exogenous perlecan. Cell proliferation was measured using a water-soluble tetrazolium dye. When cells were cultured at low densities, perlecan blocking inhibited the proliferation of VECs, and exogenous perlecan promoted the proliferation of keratinocytes. In contrast, the proliferation of fibroblasts, pre-adipocytes and vascular smooth muscle cells cultured at low densities was inhibited by exogenous perlecan. When cells were cultured at high densities, perlecan blocking promoted the proliferation of most cell types, with the exception of skeletal system-derived cells (chondrocytes and osteoblasts), which were inhibited by exogenous perlecan. Our results provide an overview of the multiple functions of perlecan in various cell types, and implicate a potential role of perlecan to inhibit undesirable activities, such as fibrosis, obesity and intimal hyperplasia.
Collapse
Affiliation(s)
- Ryosuke Nakamura
- Laboratory of Animal By-Product Science, Research Faculty of Agriculture, Hokkaido University, Sapporo, Japan
| | | | | |
Collapse
|
24
|
Ornitz DM, Itoh N. The Fibroblast Growth Factor signaling pathway. WILEY INTERDISCIPLINARY REVIEWS. DEVELOPMENTAL BIOLOGY 2015; 4:215-66. [PMID: 25772309 PMCID: PMC4393358 DOI: 10.1002/wdev.176] [Citation(s) in RCA: 1383] [Impact Index Per Article: 138.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/15/2014] [Revised: 11/23/2014] [Accepted: 01/08/2015] [Indexed: 12/13/2022]
Abstract
The signaling component of the mammalian Fibroblast Growth Factor (FGF) family is comprised of eighteen secreted proteins that interact with four signaling tyrosine kinase FGF receptors (FGFRs). Interaction of FGF ligands with their signaling receptors is regulated by protein or proteoglycan cofactors and by extracellular binding proteins. Activated FGFRs phosphorylate specific tyrosine residues that mediate interaction with cytosolic adaptor proteins and the RAS-MAPK, PI3K-AKT, PLCγ, and STAT intracellular signaling pathways. Four structurally related intracellular non-signaling FGFs interact with and regulate the family of voltage gated sodium channels. Members of the FGF family function in the earliest stages of embryonic development and during organogenesis to maintain progenitor cells and mediate their growth, differentiation, survival, and patterning. FGFs also have roles in adult tissues where they mediate metabolic functions, tissue repair, and regeneration, often by reactivating developmental signaling pathways. Consistent with the presence of FGFs in almost all tissues and organs, aberrant activity of the pathway is associated with developmental defects that disrupt organogenesis, impair the response to injury, and result in metabolic disorders, and cancer. For further resources related to this article, please visit the WIREs website.
Collapse
Affiliation(s)
- David M Ornitz
- Department of Developmental Biology, Washington University School of MedicineSt. Louis, MO, USA
- *
Correspondence to:
| | - Nobuyuki Itoh
- Graduate School of Pharmaceutical Sciences, Kyoto UniversitySakyo, Kyoto, Japan
| |
Collapse
|
25
|
Nakamura R, Nakamura F, Fukunaga S. Disruption of endogenous perlecan function improves differentiation of rat articular chondrocytesin vitro. Anim Sci J 2014; 86:449-58. [DOI: 10.1111/asj.12309] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Accepted: 07/09/2014] [Indexed: 01/11/2023]
Affiliation(s)
- Ryosuke Nakamura
- Laboratory of Animal By-Product Science; Research Faculty of Agriculture; Hokkaido University; Sapporo Japan
| | - Fumio Nakamura
- Laboratory of Animal By-Product Science; Research Faculty of Agriculture; Hokkaido University; Sapporo Japan
| | - Shigeharu Fukunaga
- Laboratory of Animal By-Product Science; Research Faculty of Agriculture; Hokkaido University; Sapporo Japan
| |
Collapse
|
26
|
Patel VN, Lombaert IMA, Cowherd SN, Shworak NW, Xu Y, Liu J, Hoffman MP. Hs3st3-modified heparan sulfate controls KIT+ progenitor expansion by regulating 3-O-sulfotransferases. Dev Cell 2014; 29:662-73. [PMID: 24960693 DOI: 10.1016/j.devcel.2014.04.024] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Revised: 04/03/2014] [Accepted: 04/22/2014] [Indexed: 11/30/2022]
Abstract
The exquisite control of growth factor function by heparan sulfate (HS) is dictated by tremendous structural heterogeneity of sulfated modifications. How specific HS structures control growth factor-dependent progenitor expansion during organogenesis is unknown. We isolated KIT+ progenitors from fetal salivary glands during a stage of rapid progenitor expansion and profiled HS biosynthetic enzyme expression. Enzymes generating a specific type of 3-O-sulfated-HS (3-O-HS) are enriched, and fibroblast growth factor 10 (FGF10)/FGF receptor 2b (FGFR2b) signaling directly regulates their expression. Bioengineered 3-O-HS binds FGFR2b and stabilizes FGF10/FGFR2b complexes in a receptor- and growth factor-specific manner. Rapid autocrine feedback increases 3-O-HS, KIT, and progenitor expansion. Knockdown of multiple Hs3st isoforms limits fetal progenitor expansion but is rescued with bioengineered 3-O-HS, which also increases adult progenitor expansion. Altering specific 3-O-sulfated epitopes provides a mechanism to rapidly respond to FGFR2b signaling and control progenitor expansion. 3-O-HS may expand KIT+ progenitors in vitro for regenerative therapy.
Collapse
Affiliation(s)
- Vaishali N Patel
- Matrix and Morphogenesis Section, Laboratory of Cell and Developmental Biology, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA
| | - Isabelle M A Lombaert
- Matrix and Morphogenesis Section, Laboratory of Cell and Developmental Biology, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA
| | - Samuel N Cowherd
- Matrix and Morphogenesis Section, Laboratory of Cell and Developmental Biology, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA
| | - Nicholas W Shworak
- Section of Cardiology, Department of Medicine, Geisel School of Medicine at Dartmouth, Hanover, NH 03756, USA
| | - Yongmei Xu
- Division of Medicinal Chemistry and Natural Products, School of Pharmacy, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Jian Liu
- Division of Medicinal Chemistry and Natural Products, School of Pharmacy, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Matthew P Hoffman
- Matrix and Morphogenesis Section, Laboratory of Cell and Developmental Biology, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
27
|
Patel VN, Hoffman MP. Salivary gland development: a template for regeneration. Semin Cell Dev Biol 2013; 25-26:52-60. [PMID: 24333774 DOI: 10.1016/j.semcdb.2013.12.001] [Citation(s) in RCA: 105] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2013] [Revised: 11/26/2013] [Accepted: 12/02/2013] [Indexed: 12/16/2022]
Abstract
The mammalian salivary gland develops as a highly branched structure designed to produce and secrete saliva. This review will focus on research on mouse submandibular gland development and the translation of this basic research toward therapy for patients suffering from salivary hypofunction. Here we review the most recent literature that has enabled a better understanding of the mechanisms of salivary gland development. Additionally, we discuss approaches proposed to restore salivary function using gene and cell-based therapy. Increasing our understanding of the developmental mechanisms involved during development is critical to design effective therapies for regeneration and repair of damaged glands.
Collapse
Affiliation(s)
- Vaishali N Patel
- Matrix and Morphogenesis Section, Laboratory of Cell and Developmental Biology, NIDCR, NIH, Bethesda, MD 20892, United States
| | - Matthew P Hoffman
- Matrix and Morphogenesis Section, Laboratory of Cell and Developmental Biology, NIDCR, NIH, Bethesda, MD 20892, United States.
| |
Collapse
|
28
|
Miaczynska M. Effects of membrane trafficking on signaling by receptor tyrosine kinases. Cold Spring Harb Perspect Biol 2013; 5:a009035. [PMID: 24186066 DOI: 10.1101/cshperspect.a009035] [Citation(s) in RCA: 93] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The intracellular trafficking machinery contributes to the spatial and temporal control of signaling by receptor tyrosine kinases (RTKs). The primary role in this process is played by endocytic trafficking, which regulates the localization of RTKs and their downstream effectors, as well as the duration and the extent of their activity. The key regulatory points along the endocytic pathway are internalization of RTKs from the plasma membrane, their sorting to degradation or recycling, and their residence in various endosomal compartments. Here I will review factors and mechanisms that modulate RTK signaling by (1) affecting receptor internalization, (2) regulating the balance between degradation and recycling of RTK, and (3) compartmentalization of signals in endosomes and other organelles. Cumulatively, these mechanisms illustrate a multilayered control of RTK signaling exerted by the trafficking machinery.
Collapse
Affiliation(s)
- Marta Miaczynska
- International Institute of Molecular and Cell Biology, Laboratory of Cell Biology, 02-109 Warsaw, Poland
| |
Collapse
|
29
|
Francavilla C, Rigbolt K, Emdal K, Carraro G, Vernet E, Bekker-Jensen D, Streicher W, Wikström M, Sundström M, Bellusci S, Cavallaro U, Blagoev B, Olsen J. Functional Proteomics Defines the Molecular Switch Underlying FGF Receptor Trafficking and Cellular Outputs. Mol Cell 2013; 51:707-22. [DOI: 10.1016/j.molcel.2013.08.002] [Citation(s) in RCA: 112] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2013] [Revised: 06/26/2013] [Accepted: 07/31/2013] [Indexed: 12/11/2022]
|
30
|
Herbert C, Schieborr U, Saxena K, Juraszek J, De Smet F, Alcouffe C, Bianciotto M, Saladino G, Sibrac D, Kudlinzki D, Sreeramulu S, Brown A, Rigon P, Herault JP, Lassalle G, Blundell TL, Rousseau F, Gils A, Schymkowitz J, Tompa P, Herbert JM, Carmeliet P, Gervasio FL, Schwalbe H, Bono F. Molecular mechanism of SSR128129E, an extracellularly acting, small-molecule, allosteric inhibitor of FGF receptor signaling. Cancer Cell 2013; 23:489-501. [PMID: 23597563 DOI: 10.1016/j.ccr.2013.02.018] [Citation(s) in RCA: 94] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2012] [Revised: 12/12/2012] [Accepted: 02/19/2013] [Indexed: 01/12/2023]
Abstract
The fibroblast growth factor (FGF)/fibroblast growth factor receptor (FGFR) signaling network plays an important role in cell growth, survival, differentiation, and angiogenesis. Deregulation of FGFR signaling can lead to cancer development. Here, we report an FGFR inhibitor, SSR128129E (SSR), that binds to the extracellular part of the receptor. SSR does not compete with FGF for binding to FGFR but inhibits FGF-induced signaling linked to FGFR internalization in an allosteric manner, as shown by crystallography studies, nuclear magnetic resonance, Fourier transform infrared spectroscopy, molecular dynamics simulations, free energy calculations, structure-activity relationship analysis, and FGFR mutagenesis. Overall, SSR is a small molecule allosteric inhibitor of FGF/FGFR signaling, acting via binding to the extracellular part of the FGFR.
Collapse
Affiliation(s)
- Corentin Herbert
- E2C and LGCR-SDI Department, Sanofi Research and Development, 31100 Toulouse, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Auciello G, Cunningham DL, Tatar T, Heath JK, Rappoport JZ. Regulation of fibroblast growth factor receptor signalling and trafficking by Src and Eps8. J Cell Sci 2012. [PMID: 23203811 DOI: 10.1242/jcs.116228] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Fibroblast growth factor receptors (FGFRs) mediate a wide spectrum of cellular responses that are crucial for development and wound healing. However, aberrant FGFR activity leads to cancer. Activated growth factor receptors undergo stimulated endocytosis, but can continue to signal along the endocytic pathway. Endocytic trafficking controls the duration and intensity of signalling, and growth factor receptor signalling can lead to modifications of trafficking pathways. We have developed live-cell imaging methods for studying FGFR dynamics to investigate mechanisms that coordinate the interplay between receptor trafficking and signal transduction. Activated FGFR enters the cell following recruitment to pre-formed clathrin-coated pits (CCPs). However, FGFR activation stimulates clathrin-mediated endocytosis; FGF treatment increases the number of CCPs, including those undergoing endocytosis, and this effect is mediated by Src and its phosphorylation target Eps8. Eps8 interacts with the clathrin-mediated endocytosis machinery and depletion of Eps8 inhibits FGFR trafficking and immediate Erk signalling. Once internalized, FGFR passes through peripheral early endosomes en route to recycling and degredative compartments, through an Src- and Eps8-dependent mechanism. Thus Eps8 functions as a key coordinator in the interplay between FGFR signalling and trafficking. This work provides the first detailed mechanistic analysis of growth factor receptor clustering at the cell surface through signal transduction and endocytic trafficking. As we have characterised the Src target Eps8 as a key regulator of FGFR signalling and trafficking, and identified the early endocytic system as the site of Eps8-mediated effects, this work provides novel mechanistic insight into the reciprocal regulation of growth factor receptor signalling and trafficking.
Collapse
Affiliation(s)
- Giulio Auciello
- CRUK Growth Factor Group, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| | | | | | | | | |
Collapse
|
32
|
Fang X, Bai C, Wang X. Potential clinical application of KGF-2 (FGF-10) for acute lung injury/acute respiratory distress syndrome. Expert Rev Clin Pharmacol 2012; 3:797-805. [PMID: 22111782 DOI: 10.1586/ecp.10.59] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Acute lung injury (ALI)/acute respiratory distress syndrome (ARDS) is an acute life-threatening form of hypoxemic respiratory failure with a high mortality rate, and there is still a great need for more effective therapies for such a severe and lethal disease. Dysfunction of endothelial and epithelial barriers is one of the most important mechanisms in hypoxia-associated ALI/ARDS. The acceleration of the epithelial repair process in the injured lung may provide an effective therapeutic target. KGF-2, a potent alveolar epithelial cell mitogen, plays an important role in organ morphogenesis and epithelial differentiation, and modulates a variety of mechanisms recognized to be important in alveolar repair and resolution in ALI/ARDS. Preclinical and clinical studies have suggested that KGF-2 may be the candidate of novel therapies for alveolar epithelial damage during ALI/ARDS.
Collapse
Affiliation(s)
- Xiaocong Fang
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China.
| | | | | |
Collapse
|
33
|
Cholesteatoma-associated fibroblasts modulate epithelial growth and differentiation through KGF/FGF7 secretion. Histochem Cell Biol 2012; 138:251-69. [PMID: 22481617 PMCID: PMC3407559 DOI: 10.1007/s00418-012-0947-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/22/2012] [Indexed: 11/04/2022]
Abstract
The keratinocyte growth factor (KGF/FGF7), produced by stromal cells, is a key paracrine mediator of epithelial proliferation, differentiation and migration. Expression of the growth factor is increased in wound healing and in hyperproliferative epithelial diseases, as a consequence of the activation of dermal fibroblasts by the inflammatory microenvironment. The middle ear cholesteatoma, an aural epidermal pathology characterized by keratinocyte hyperproliferation and chronic inflammation, may represent a model condition to study the epithelial-mesenchymal interactions. To develop an in vitro model for this disease, we isolated and characterized human primary cultures of fibroblasts associated with the cholesteatoma lesion, analyzing their secretory behaviour and degree of differentiation or activation. Compared to the perilesional or control normal fibroblasts, all cultures derived from cholesteatoma tissues were less proliferating and more differentiated and their highly variable activated phenotype correlated with the secretion of KGF as well as of metalloproteases 2 and 9. Culture supernatants collected from the cholesteatoma-associated fibroblasts were able to increase the proliferation and differentiation of human keratinocytes assessed by the expression of Ki67 and keratin-1 markers. The single crucial contribution of the KGF released by fibroblasts on the keratinocyte biological response was shown by the specific, although partial, block induced by inhibiting the KGF receptor or by immunoneutralizing the growth factor. Altogether, these results suggest that the activation of the stromal fibroblasts present in the pathological tissue, and the consequent increased secretion of KGF, play a crucial role in the deregulation of the epidermal proliferation and differentiation.
Collapse
|
34
|
Polarized endocytosis of the keratinocyte growth factor receptor in migrating cells: role of SRC-signaling and cortactin. PLoS One 2011; 6:e29159. [PMID: 22195012 PMCID: PMC3237600 DOI: 10.1371/journal.pone.0029159] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2011] [Accepted: 11/22/2011] [Indexed: 11/19/2022] Open
Abstract
Cell migration is a physiological process that requires endocytic trafficking and polarization of adhesion molecules and receptor tyrosine kinases (RTKs) to the leading edge. Many growth factors are able to induce motility by binding to specific RTK on target cells. Among them, keratinocyte growth factor (KGF or FGF7) and fibroblast growth factor 10 (FGF10), members of the FGF family, are motogenic for keratinocytes, and exert their action by binding to the keratinocyte growth factor receptor (KGFR), a splicing variant of FGFR2, exclusively expressed on epithelial cells. Here we analyzed the possible role of cortactin, an F-actin binding protein which is tyrosine phosphorylated by Src and is involved in KGFR-mediated cell migration, in the KGFR endocytosis and polarization to the leading edge of migrating cells upon ligand-induced stimulation. Biochemical phosphorylation study revealed that both KGF and FGF10 were able to induce tyrosine phosphorylation of Src and in turn of cortactin, as demonstrated by using the specific pharmacological Src-inhibitor SU6656, although FGF10 effect was delayed with respect to that promoted by KGF. Immunofluorescence analysis demonstrated the polarized localization of KGFR upon ligand stimulation to the leading edge of migrating keratinocytes, process that was regulated by Src. Moreover, we showed that the colocalization of cortactin with KGFR at the plasma membrane protrusions and on early endosomes after KGF and FGF10 treatment was Src-dependent. Further, by using a RNA interference approach through microinjection, we showed that cortactin is required for KGFR endocytosis and that the clathrin-dependent internalization of the receptor is a critical event for its polarization. Finally, KGFR expression and polarization enhanced cell migration in a scratch assay. Our results indicate that both Src and cortactin play a key role in the KGFR endocytosis and polarization at the leading edge of migrating keratinocytes, supporting the crucial involvement of RTK trafficking in cell motility.
Collapse
|
35
|
The receptor tyrosine kinase FGFR2b/KGFR controls early differentiation of human keratinocytes. PLoS One 2011; 6:e24194. [PMID: 21957444 PMCID: PMC3177842 DOI: 10.1371/journal.pone.0024194] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2011] [Accepted: 08/04/2011] [Indexed: 12/23/2022] Open
Abstract
The FGFRs trigger divergent responses, such as proliferation and differentiation, and the cell type as well as the context-dependent signaling are crucial for the functional outcome. The FGFR2b/KGFR is expressed exclusively on epithelial cells and plays a key role in skin homeostasis. Here we analyzed in vitro the role of KGFR in the early differentiation of keratinocytes modulating its expression by KGFR cDNA transient transfection or KGFR siRNA microinjection and inducing a synchronous wave of differentiation in pre-confluent cells. Immunofluorescence, biochemical and molecular approaches demonstrated that KGFR overexpression increased the early differentiation marker keratin 1 at both transcriptional and translational levels, while receptor depletion reduced it. Ligand-dependent receptor activation and signaling were required for this differentiative effect. Overexpression of kinase negative KGFR mutant or Tyr769 KGFR signaling mutant, which is not able to recruit and activate PLC-γ, showed that the receptor kinase activity, but not its PLCγ-mediated signaling, is required for differentiation. Reduction of K1 expression, obtained by AKT inhibition, demonstrated that the PI3K/Akt signaling pathway is involved in the control of KGFR-mediated keratinocyte differentiation. This in vitro experimental model indicates that FGFR2b/KGFR expression represents a key event regulating keratinocyte early differentiation during the switch from undifferentiated to differentiating cells.
Collapse
|
36
|
Abstract
FGFs (fibroblast growth factors) and their receptors (FGFRs) play essential roles in tightly regulating cell proliferation, survival, migration and differentiation during development and adult life. Deregulation of FGFR signalling, on the other hand, has been associated with many developmental syndromes, and with human cancer. In cancer, FGFRs have been found to become overactivated by several mechanisms, including gene amplification, chromosomal translocation and mutations. FGFR alterations are detected in a variety of human cancers, such as breast, bladder, prostate, endometrial and lung cancers, as well as haematological malignancies. Accumulating evidence indicates that FGFs and FGFRs may act in an oncogenic fashion to promote multiple steps of cancer progression by inducing mitogenic and survival signals, as well as promoting epithelial-mesenchymal transition, invasion and tumour angiogenesis. Therapeutic strategies targeting FGFs and FGFRs in human cancer are therefore currently being explored. In the present review we will give an overview of FGF signalling, the main FGFR alterations found in human cancer to date, how they may contribute to specific cancer types and strategies for therapeutic intervention.
Collapse
|
37
|
Degnin CR, Laederich MB, Horton WA. Ligand activation leads to regulated intramembrane proteolysis of fibroblast growth factor receptor 3. Mol Biol Cell 2011; 22:3861-73. [PMID: 21865593 PMCID: PMC3192865 DOI: 10.1091/mbc.e11-01-0080] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
FGFR3 is implicated in several human diseases. Following activation and endocytosis, FGFR3 undergoes sequential ectodomain and intramembrane cleavages to generate a soluble cytoplasmic fragment that can translocate to the nucleus. Fibroblast growth factor receptor 3 (FGFR3) is a major negative regulator of bone growth that inhibits the proliferation and differentiation of growth plate chondrocytes. Activating mutations of its c isoform cause dwarfism in humans; somatic mutations can drive oncogenic transformation in multiple myeloma and bladder cancer. How these distinct activities arise is not clear. FGFR3 was previously shown to undergo proteolytic cleavage in the bovine rib growth plate, but this was not explored further. Here, we show that FGF1 induces regulated intramembrane proteolysis (RIP) of FGFR3. The ectodomain is proteolytically cleaved (S1) in response to ligand-induced receptor activation, but unlike most RIP target proteins, it requires endocytosis and does not involve a metalloproteinase. S1 cleavage generates a C-terminal domain fragment that initially remains anchored in the membrane, is phosphorylated, and is spatially distinct from the intact receptor. Ectodomain cleavage is followed by intramembrane cleavage (S2) to generate a soluble intracellular domain that is released into the cytosol and can translocate to the nucleus. We identify the S1 cleavage site and show that γ-secretase mediates the S2 cleavage event. In this way we demonstrate a mechanism for the nuclear localization of FGFR3 in response to ligand activation, which may occur in both development and disease.
Collapse
Affiliation(s)
- Catherine R Degnin
- Research Center, Shriners Hospital for Children, Portland, OR 97239, USA
| | | | | |
Collapse
|
38
|
Haugsten EM, Zakrzewska M, Brech A, Pust S, Olsnes S, Sandvig K, Wesche J. Clathrin- and dynamin-independent endocytosis of FGFR3--implications for signalling. PLoS One 2011; 6:e21708. [PMID: 21779335 PMCID: PMC3136467 DOI: 10.1371/journal.pone.0021708] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2011] [Accepted: 06/06/2011] [Indexed: 11/20/2022] Open
Abstract
Endocytosis of tyrosine kinase receptors can influence both the duration and the specificity of the signal emitted. We have investigated the mechanisms of internalization of fibroblast growth factor receptor 3 (FGFR3) and compared it to that of FGFR1 which is internalized predominantly through clathrin-mediated endocytosis. Interestingly, we observed that FGFR3 was internalized at a slower rate than FGFR1 indicating that it may use a different endocytic mechanism than FGFR1. Indeed, after depletion of cells for clathrin, internalization of FGFR3 was only partly inhibited while endocytosis of FGFR1 was almost completely abolished. Similarly, expression of dominant negative mutants of dynamin resulted in partial inhibition of the endocytosis of FGFR3 whereas internalization of FGFR1 was blocked. Interfering with proposed regulators of clathrin-independent endocytosis such as Arf6, flotillin 1 and 2 and Cdc42 did not affect the endocytosis of FGFR1 or FGFR3. Furthermore, depletion of clathrin decreased the degradation of FGFR1 resulting in sustained signalling. In the case of FGFR3, both the degradation and the signalling were only slightly affected by clathrin depletion. The data indicate that clathrin-mediated endocytosis is required for efficient internalization and downregulation of FGFR1 while FGFR3, however, is internalized by both clathrin-dependent and clathrin-independent mechanisms.
Collapse
MESH Headings
- Blotting, Western
- Cell Line, Tumor
- Cell Movement/genetics
- Cell Movement/physiology
- Clathrin/genetics
- Clathrin/metabolism
- Dynamins/genetics
- Dynamins/metabolism
- Endocytosis/genetics
- Endocytosis/physiology
- Humans
- Microscopy, Confocal
- Receptor, Fibroblast Growth Factor, Type 1/genetics
- Receptor, Fibroblast Growth Factor, Type 1/metabolism
- Receptor, Fibroblast Growth Factor, Type 3/genetics
- Receptor, Fibroblast Growth Factor, Type 3/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
- Signal Transduction/genetics
- Signal Transduction/physiology
Collapse
Affiliation(s)
- Ellen Margrethe Haugsten
- Centre for Cancer Biomedicine, Faculty Division Norwegian Radium Hospital, University of Oslo, Oslo, Norway
- Department of Biochemistry, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo, Norway
| | - Malgorzata Zakrzewska
- Centre for Cancer Biomedicine, Faculty Division Norwegian Radium Hospital, University of Oslo, Oslo, Norway
- Department of Biochemistry, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo, Norway
- Faculty of Biotechnology, University of Wroclaw, Wroclaw, Poland
| | - Andreas Brech
- Centre for Cancer Biomedicine, Faculty Division Norwegian Radium Hospital, University of Oslo, Oslo, Norway
- Department of Biochemistry, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo, Norway
| | - Sascha Pust
- Centre for Cancer Biomedicine, Faculty Division Norwegian Radium Hospital, University of Oslo, Oslo, Norway
- Department of Biochemistry, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo, Norway
| | - Sjur Olsnes
- Centre for Cancer Biomedicine, Faculty Division Norwegian Radium Hospital, University of Oslo, Oslo, Norway
- Department of Biochemistry, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo, Norway
| | - Kirsten Sandvig
- Centre for Cancer Biomedicine, Faculty Division Norwegian Radium Hospital, University of Oslo, Oslo, Norway
- Department of Biochemistry, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo, Norway
| | - Jørgen Wesche
- Centre for Cancer Biomedicine, Faculty Division Norwegian Radium Hospital, University of Oslo, Oslo, Norway
- Department of Biochemistry, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo, Norway
- * E-mail:
| |
Collapse
|
39
|
Dabrowski A, Umemori H. Orchestrating the synaptic network by tyrosine phosphorylation signalling. J Biochem 2011; 149:641-53. [PMID: 21508038 PMCID: PMC3143439 DOI: 10.1093/jb/mvr047] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2011] [Accepted: 04/08/2011] [Indexed: 01/07/2023] Open
Abstract
The establishment of a functional brain requires coordinated and stereotyped formation of synapses between neurons. For this, trans-synaptic molecular cues (synaptic organizers) are exchanged between a neuron and its target to organize appropriate synapses. The understanding of signalling mechanisms by which such synaptic organizers lead to synapse formation is just being elucidated. However, recent studies revealed that some of these cues act through receptor protein tyrosine kinases (RPTKs) or phosphatases (RPTPs). Synaptogenic RPTKs and RPTPs pattern synaptic network through affecting local protein-protein binding dynamics, changing the phosphorylation state of signalling cascades, or promoting gene expression. Each RPTK or RPTP has distinct roles in synapse formation, serving at different synapses or showing differential synaptogenic effects. Thus, tyrosine phosphorylation signalling plays critical roles in building the orchestrated synaptic circuitry in the brain.
Collapse
Affiliation(s)
- Ania Dabrowski
- Molecular & Behavioral Neuroscience Institute, Medical Scientist Training Program, Neuroscience Graduate Program and Department of Biological Chemistry, University of Michigan Medical School, Ann Arbor, MI 48109-2200, USA
| | - Hisashi Umemori
- Molecular & Behavioral Neuroscience Institute, Medical Scientist Training Program, Neuroscience Graduate Program and Department of Biological Chemistry, University of Michigan Medical School, Ann Arbor, MI 48109-2200, USA
| |
Collapse
|
40
|
Belleudi F, Leone L, Purpura V, Cannella F, Scrofani C, Torrisi MR. HPV16 E5 affects the KGFR/FGFR2b-mediated epithelial growth through alteration of the receptor expression, signaling and endocytic traffic. Oncogene 2011; 30:4963-76. [PMID: 21625213 DOI: 10.1038/onc.2011.203] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The E5 oncoprotein of the human papillomavirus type 16 (HPV16 E5) cooperates in cervical carcinogenesis and in epithelial transformation deregulating cell growth, survival and differentiation through the modulation of growth factor receptors. Among the epithelial receptor tyrosine kinases, the keratinocyte growth factor receptor/fibroblast growth factor receptor 2b (KGFR/FGFR2b) is a major paracrine mediator of epithelial homeostasis and appears to have an unique and unusual role in epithelial tissues, exerting a tumor-suppressive function in vitro and in vivo. With the aim to better elucidate the molecular events involved in the pathological activity of 16E5, we investigated if the viral protein would be able to affect the KGFR expression, signaling and turnover by interference with its degradative and recycling endocytic pathways. Quantitative reverse transcriptase-PCR and biochemical approaches on human keratinocytes transfected with 16E5-HA showed that E5 protein is able to induce KGFR down-modulation at both transcript and protein levels. Immunofluorescence microscopy in double-transfected cells expressing both E5 and KGFR revealed that the viral protein alters the receptor endocytic trafficking and triggers its endosomal sorting to the indirect juxtanuclear recycling pathway. The shift from lysosomal degradation to recycling at the plasma membrane correlates with a reduced phosphorylation of the fibroblast growth factor receptor substrate-2α tyrosine 196, the major docking site for Grb2-Cbl complexes responsible for receptor ubiquitination and degradation. 5'-Bromo-deoxyuridine incorporation assay demonstrated that expression of 16E5 induces a decrease in the growth response to the receptor ligands as a consequence of KGFR down-modulation, suggesting that 16E5 might have a role on HPV infection in perturbing the KGFR-mediated physiological behavior of confluent keratinocytes committed to differentiation.
Collapse
Affiliation(s)
- F Belleudi
- Istituto Pasteur-Fondazione Cenci Bolognetti, Dipartimento di Medicina Clinica e Molecolare, Sapienza Università di Roma, Rome, Italy
| | | | | | | | | | | |
Collapse
|
41
|
Inhibition of fibroblast growth factor receptor 1 endocytosis promotes axonal branching of adult sensory neurons. Neuroscience 2011; 188:13-22. [PMID: 21575685 DOI: 10.1016/j.neuroscience.2011.04.064] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2011] [Revised: 04/26/2011] [Accepted: 04/27/2011] [Indexed: 01/21/2023]
Abstract
Fibroblast growth factors (FGFs) promote axon growth during development and regeneration of the nervous system. Among the four types of FGF receptors (FGFRs), FGFR1 is expressed in adult sensory neurons of dorsal root ganglia (DRG), and overexpression of FGFR1 promotes FGF-2-induced elongative axon growth in vitro. Ligand-induced activation of FGFR1 is followed by endocytosis and lysosomal degradation, which leads to the termination of receptor signaling. We previously reported that the lysosomal inhibitor leupeptin enhances FGF-2-induced elongative axon growth of adult DRG neurons overexpressing FGFR1. To better understand the role of subcellular localization of FGFR1 in axon growth, we analyzed the effects of inhibition of endocytosis of FGFR1 on FGF-2-induced neurite outgrowth in PC12 pheochromocytoma cells and adult DRG neurons. The endocytosis inhibitors methyl-β-cyclodextrin (MβCD) and chlorpromazine enhanced surface localization of FGFR1 in PC12 cells and DRG neurons. Furthermore, MβCD and chlorpromazine increased FGF-2-induced neurite outgrowth of PC12 cells and axonal branching of adult DRG neurons overexpressing FGFR1, whereas MβCD inhibited FGF-2-induced axonal elongation. Analysis of the signaling pathways involved in axon morphology revealed that FGF-2-induced phosphorylation of extracellular signal-regulated kinase (ERK) and Akt was increased by inhibition of FGFR1 endocytosis. Together, our results imply that inhibition of FGFR1 endocytosis by MβCD or chlorpromazine promotes FGF-2-induced axonal branching. The results of this study confirm that internalization of FGFR1 controls axon growth and morphology of adult sensory neurons via selective activation of intracellular signaling pathways.
Collapse
|
42
|
Jean S, Mikryukov A, Tremblay MG, Baril J, Guillou F, Bellenfant S, Moss T. Extended-synaptotagmin-2 mediates FGF receptor endocytosis and ERK activation in vivo. Dev Cell 2010; 19:426-39. [PMID: 20833364 DOI: 10.1016/j.devcel.2010.08.007] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2009] [Revised: 06/14/2010] [Accepted: 07/14/2010] [Indexed: 10/19/2022]
Abstract
Targeting of activated plasma membrane receptors to endocytic pathways is important in determining the outcome of growth factor signaling. However, the molecular mechanisms are still poorly understood. Here, we show that the synaptotagmin-related membrane protein E-Syt2 is essential for rapid endocytosis of the activated FGF receptor and for functional signal transduction during Xenopus development. E-Syt2 depletion prevents an early phase of activated FGF receptor endocytosis that we show is required for ERK activation and the induction of the mesoderm. E-Syt2 interacts selectively with the activated FGF receptor and with Adaptin-2, and is required upstream of Ras activation and of receptor autophosphorylation for ERK activation and the induction of the mesodermal marker Xbra. The data identify E-Syt2 as an endocytic adaptor for the clathrin-mediated pathway whose function is conserved in human and suggest a broader role for the E-Syt subfamily in growth factor signaling.
Collapse
Affiliation(s)
- Steve Jean
- Cancer Research Centre and Department of Medical Biology, Medical Biochemistry and Pathology, Laval University, Hôtel-Dieu de Québec, 9 rue McMahon, G1R 2J6 Québec, Canada
| | | | | | | | | | | | | |
Collapse
|
43
|
Belleudi F, Purpura V, Scrofani C, Persechino F, Leone L, Torrisi MR. Expression and signaling of the tyrosine kinase FGFR2b/KGFR regulates phagocytosis and melanosome uptake in human keratinocytes. FASEB J 2010; 25:170-81. [PMID: 20844240 DOI: 10.1096/fj.10-162156] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Membrane and actin cytoskeleton dynamics during phagocytosis can be triggered and amplified by the signal transduction of receptor tyrosine kinases. The epidermal keratinocytes appear to use the phagocytic mechanism of uptake to ingest melanosomes released by the melanocytes and play a pivotal role in the transfer process. We have previously demonstrated that the keratinocyte growth factor KGF/FGF7 promotes the melanosome uptake through activation of its receptor tyrosine kinase FGFR2b/KGFR. The aim of the present study was to investigate the contribution of KGFR expression, activation, and signaling in regulating the phagocytic process and the melanosome transfer. Phagocytosis was analyzed in vitro using fluorescent latex beads on human keratinocytes induced to differentiate. Melanosome transfer was investigated in keratinocyte-melanocyte cocultures. KGFR depletion by small interfering RNA microinjection and overexpression by transfection of wild type or defective mutant KGFR were performed to demonstrate the direct effect of the receptor on phagocytosis and melanosome transfer. Colocalization of the phagocytosed beads with the internalized receptors in phagolysosomes was analyzed by optical sectioning and 3-dimensional reconstruction. KGFR ligands triggered phagocytosis and melanosome transfer in differentiated keratinocytes, and receptor kinase activity and signaling were required for these effects, suggesting that FGFR2b/KGFR expression/activity and PLCγ signaling pathway play crucial roles in phagocytosis.
Collapse
Affiliation(s)
- Francesca Belleudi
- Istituto Pasteur-Fondazione Cenci Bolognetti, Dipartimento di Medicina Clinica e Molecolare, Sapienza Università di Roma, Rome, Italy
| | | | | | | | | | | |
Collapse
|
44
|
Vinsonneau C, Girshovich A, M'rad MB, Perez J, Mesnard L, Vandermersch S, Placier S, Letavernier E, Baud L, Haymann JP. Intrarenal urothelium proliferation: an unexpected early event following ischemic injury. Am J Physiol Renal Physiol 2010; 299:F479-86. [PMID: 20591940 DOI: 10.1152/ajprenal.00585.2009] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Identification of renal cell progenitors and recognition of the events contributing to cell regeneration following ischemia-reperfusion injury (IRI) are a major challenge. In a mouse model of unilateral renal IRI, we demonstrated that the first cells to proliferate within injured kidneys were urothelial cells expressing the progenitor cell marker cytokeratin 14. A systematic cutting of the injured kidney revealed that these urothelial cells were located in the deep cortex at the corticomedullary junction in the vicinity of lobar vessels. Contrary to multilayered bladder urothelium, these intrarenal urothelial cells located in the upper part of the medulla constitute a monolayered barrier and express among uroplakins only uroplakin III. However, like bladder progenitors, intrarenal urothelial cells proliferated through a FGF receptor-2 (FGFR2)-mediated process. They strongly expressed FGFR2 and proliferated in vivo after recombinant FGF7 administration to control mice. In addition, IRI led to FGFR phosphorylation together with the selective upregulation of FGF7 and FGF2. Conversely, by day 2 following IRI, renal urothelial cell proliferation was significantly inhibited by FGFR2 antisense oligonucleotide administration into an intrarenal urinary space. Of notice, no significant migration of these early dividing urothelial cells was detected in the cortex within 7 days following IRI. Thus our data show that following IRI, proliferation of urothelial cells is mediated by the FGFR2 pathway and precedes tubular cell proliferation, indicating a particular sensitivity of this structure to changes caused by the ischemic process.
Collapse
Affiliation(s)
- C Vinsonneau
- INSERM UMRS, Université Pierre et Marie Curie Paris, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Hille A, Grüger S, Christiansen H, Wolff HA, Volkmer B, Lehmann J, Dörr W, Rave-Fränk M. Effect of tumour-cell-derived or recombinant keratinocyte growth factor (KGF) on proliferation and radioresponse of human epithelial tumour cells (HNSCC) and normal keratinocytes in vitro. RADIATION AND ENVIRONMENTAL BIOPHYSICS 2010; 49:261-270. [PMID: 20213138 PMCID: PMC2855434 DOI: 10.1007/s00411-010-0271-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/27/2009] [Accepted: 02/05/2010] [Indexed: 05/28/2023]
Abstract
Purpose of this work was to test the effect of tumour-cell-derived keratinocyte growth factor (KGF) or recombinant KGF (palifermin) on cell proliferation and radiation response of human HNSCC cells and normal keratinocytes in vitro. Four tumour cell cultures derived from head and neck squamous cell carcinomas, primary keratinocytes, and immortalized keratinocytes were analysed. Fibroblasts, the natural source of KGF protein, served as controls. KGF expression was observed in primary and immortalized keratinocytes, fibroblasts, and in tumour cells, while significant KGF receptor expression was only found in keratinocytes. Recombinant KGF as well as tumour-cell-derived KGF caused a significant growth stimulation and radioprotection in keratinocytes, which was abolished by a neutralizing anti-KGF antibody. This indicates that tumour-cell-derived KGF is biologically active. In the tumour cell lines, no significant growth stimulation was induced by recombinant KGF, and the neutralizing antibody did not influence tumour cell growth or radiation response. Our results indicate that the normal, paracrine KGF regulatory mechanisms, which are based on KGF receptor expression, are lost in malignant cells, with the consequence of irresponsiveness of the tumour cells to exogenous KGF. In face of the amelioration of the radiation response of normal epithelia, demonstrated in various clinical and various preclinical animal studies, recombinant KGF represents a candidate for the selective protection of normal epithelia during radio(chemo) therapy of squamous cell carcinoma.
Collapse
Affiliation(s)
- Andrea Hille
- Department of Radiotherapy and Radiooncology, University Medicine Göttingen, Göttingen, Germany
| | - Susanne Grüger
- Department of Radiotherapy and Radiooncology, University Medicine Göttingen, Göttingen, Germany
| | - Hans Christiansen
- Department of Radiotherapy and Radiooncology, University Medicine Göttingen, Göttingen, Germany
| | - Hendrik A. Wolff
- Department of Radiotherapy and Radiooncology, University Medicine Göttingen, Göttingen, Germany
| | - Beate Volkmer
- Dermatology Centre Elbeklinikum Buxtehude, Buxtehude, Germany
| | - Jörg Lehmann
- Department of Radiation Oncology, University of California Davis School of Medicine, Sacramento, CA USA
| | - Wolfgang Dörr
- Department of Radiotherapy and Radiation Oncology, Medical Faculty Carl Gustav Carus, University of Technology, Dresden, Germany
| | - Margret Rave-Fränk
- Department of Radiotherapy and Radiooncology, University Medicine Göttingen, Göttingen, Germany
| |
Collapse
|
46
|
Chanut-Delalande H, Jung AC, Baer MM, Lin L, Payre F, Affolter M. The Hrs/Stam complex acts as a positive and negative regulator of RTK signaling during Drosophila development. PLoS One 2010; 5:e10245. [PMID: 20422006 PMCID: PMC2858154 DOI: 10.1371/journal.pone.0010245] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2009] [Accepted: 03/30/2010] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Endocytosis is a key regulatory step of diverse signalling pathways, including receptor tyrosine kinase (RTK) signalling. Hrs and Stam constitute the ESCRT-0 complex that controls the initial selection of ubiquitinated proteins, which will subsequently be degraded in lysosomes. It has been well established ex vivo and during Drosophila embryogenesis that Hrs promotes EGFR down regulation. We have recently isolated the first mutations of stam in flies and shown that Stam is required for air sac morphogenesis, a larval respiratory structure whose formation critically depends on finely tuned levels of FGFR activity. This suggest that Stam, putatively within the ESCRT-0 complex, modulates FGF signalling, a possibility that has not been examined in Drosophila yet. PRINCIPAL FINDINGS Here, we assessed the role of the Hrs/Stam complex in the regulation of signalling activity during Drosophila development. We show that stam and hrs are required for efficient FGFR signalling in the tracheal system, both during cell migration in the air sac primordium and during the formation of fine cytoplasmic extensions in terminal cells. We find that stam and hrs mutant cells display altered FGFR/Btl localisation, likely contributing to impaired signalling levels. Electron microscopy analyses indicate that endosome maturation is impaired at distinct steps by hrs and stam mutations. These somewhat unexpected results prompted us to further explore the function of stam and hrs in EGFR signalling. We show that while stam and hrs together downregulate EGFR signalling in the embryo, they are required for full activation of EGFR signalling during wing development. CONCLUSIONS/SIGNIFICANCE Our study shows that the ESCRT-0 complex differentially regulates RTK signalling, either positively or negatively depending on tissues and developmental stages, further highlighting the importance of endocytosis in modulating signalling pathways during development.
Collapse
Affiliation(s)
- Hélène Chanut-Delalande
- Biozentrum der Universität Basel, Abteilung Zellbiologie, Basel, Switzerland
- Université de Toulouse, UPS, Centre de Biologie du Développement, Université Paul Sabatier, Toulouse, France
- CNRS, UMR5547, Centre de Biologie du Développement, Toulouse, France
| | - Alain C. Jung
- Biozentrum der Universität Basel, Abteilung Zellbiologie, Basel, Switzerland
| | - Magdalena M. Baer
- Biozentrum der Universität Basel, Abteilung Zellbiologie, Basel, Switzerland
| | - Li Lin
- Biozentrum der Universität Basel, Abteilung Zellbiologie, Basel, Switzerland
| | - François Payre
- Université de Toulouse, UPS, Centre de Biologie du Développement, Université Paul Sabatier, Toulouse, France
- CNRS, UMR5547, Centre de Biologie du Développement, Toulouse, France
| | - Markus Affolter
- Biozentrum der Universität Basel, Abteilung Zellbiologie, Basel, Switzerland
| |
Collapse
|
47
|
Gu J, Faundez V, Werner E. Endosomal recycling regulates Anthrax Toxin Receptor 1/Tumor Endothelial Marker 8-dependent cell spreading. Exp Cell Res 2010; 316:1946-57. [PMID: 20382142 DOI: 10.1016/j.yexcr.2010.03.026] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2010] [Revised: 03/30/2010] [Accepted: 03/31/2010] [Indexed: 11/26/2022]
Abstract
Mechanisms for receptor-mediated anthrax toxin internalization and delivery to the cytosol are well understood. However, far less is known about the fate followed by anthrax toxin receptors prior and after cell exposure to the toxin. We report that Anthrax Toxin Receptor 1/Tumor Endothelial Marker 8 (TEM8) localized at steady state in Rab11a-positive and transferrin receptor-containing recycling endosomes. TEM8 followed a slow constitutive recycling route of approximately 30min as determined by pulsed surface biotinylation and chase experiments. A Rab11a dominant negative mutant and Myosin Vb tail expression impaired TEM8 recycling by sequestering TEM8 in intracellular compartments. Sequestration of TEM8 in intracellular compartments with monensin coincided with increased TEM8 association with a multi-protein complex isolated with antibodies against transferrin receptor. Addition of the cell-binding component of anthrax toxin, Protective Antigen, reduced TEM8 half-life from 7 to 3 hours, without preventing receptor recycling. Pharmacological and molecular perturbation of recycling endosome function using monensin, dominant negative Rab11a, or myosin Vb tail, reduced PA binding efficiency and TEM8-dependent cell spreading on PA-coated surfaces without affecting toxin delivery to the cytosol. These results indicate that the intracellular fate of TEM8 differentially affect its cell adhesion and cell intoxication functions.
Collapse
Affiliation(s)
- Jingsheng Gu
- Department of Cell Biology, Emory University School of Medicine, Atlanta, Georgia 30322, USA
| | | | | |
Collapse
|
48
|
Melnik BC, Schmitz G, Zouboulis CC. Anti-Acne Agents Attenuate FGFR2 Signal Transduction in Acne. J Invest Dermatol 2009; 129:1868-77. [DOI: 10.1038/jid.2009.8] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
|
49
|
Belleudi F, Leone L, Maggio M, Torrisi MR. Hrs regulates the endocytic sorting of the fibroblast growth factor receptor 2b. Exp Cell Res 2009; 315:2181-91. [DOI: 10.1016/j.yexcr.2009.03.022] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2009] [Revised: 03/27/2009] [Accepted: 03/29/2009] [Indexed: 11/28/2022]
|
50
|
Melnik BC. Role of FGFR2-signaling in the pathogenesis of acne. DERMATO-ENDOCRINOLOGY 2009; 1:141-56. [PMID: 20436882 PMCID: PMC2835907 DOI: 10.4161/derm.1.3.8474] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/15/2009] [Accepted: 03/18/2009] [Indexed: 01/10/2023]
Abstract
It is the purpose of this review to extend our understanding of the fibroblast growth factor (FGF) receptor-2b-signaling network in the pathogenesis of acne. A new concept of the role of FGFR2b-signaling in dermal-epithelial interaction for skin appendage formation, pilosebaceous follicle homeostasis, comedogenesis, sebaceous gland proliferation and lipogenesis is presented. The FGFR2-gain-of-function mutations in Apert syndrome and unilateral acneiform nevus are most helpful model diseases pointing the way to androgen-dependent dermalepithelial FGFR2-signaling in acne. Androgen-mediated upregulation of FGFR2b-signaling in acne-prone skin appears to be involved in the pathogenesis of acne vulgaris. In organotypic skin cultures, keratinocyte-derived interleukin-1alpha stimulated fibroblasts to secrete FGF7 which stimulated FGFR2b-mediated keratinocyte proliferation. Postnatal deletion of FGFR2b in mice resulted in severe sebaceous gland atrophy. The importance of FGFR2b in sebaceous gland physiology is further supported by the mode of action of anti-acne agents which have been proposed to attenuate FGFR2b-signaling. Downregulation of FGFR2b-signaling by isotretinoin explains its therapeutic effect in acne. Downregulation of FGFR2b-signaling during the first trimester of pregnancy disturbs branched morphogenesis and explains retinoid embryotoxicity. Insulin-like growth factor-1 (IGF-1), the mediator of growth hormone during puberty, intracts with androgen-dependent FGFR2b-signaling and links androgen- and FGF-mediated signal transduction important in sebaceous gland homeostasis. The search for a follicular defect in the dermalepithelial regulation of growth factor-signaling in acne-prone skin appears to be a most promising approach to clarify the pathogenesis of acne.
Collapse
Affiliation(s)
- Bodo C Melnik
- Department of Dermatology; Environmental Medicine and Health Theory; University of Osnabrück; Germany
| |
Collapse
|