1
|
Abstract
The endothelium is a dynamic, semipermeable layer lining all blood vessels, regulating blood vessel formation and barrier function. Proper composition and function of the endothelial barrier are required for fluid homeostasis, and clinical conditions characterized by barrier disruption are associated with severe morbidity and high mortality rates. Endothelial barrier properties are regulated by cell-cell junctions and intracellular signaling pathways governing the cytoskeleton, but recent insights indicate an increasingly important role for integrin-mediated cell-matrix adhesion and signaling in endothelial barrier regulation. Here, we discuss diseases characterized by endothelial barrier disruption, and provide an overview of the composition of endothelial cell-matrix adhesion complexes and associated signaling pathways, their crosstalk with cell-cell junctions, and with other receptors. We further present recent insights into the role of cell-matrix adhesions in the developing and mature/adult endothelium of various vascular beds, and discuss how the dynamic regulation and turnover of cell-matrix adhesions regulates endothelial barrier function in (patho)physiological conditions like angiogenesis, inflammation and in response to hemodynamic stress. Finally, as clinical conditions associated with vascular leak still lack direct treatment, we focus on how understanding of endothelial cell-matrix adhesion may provide novel targets for treatment, and discuss current translational challenges and future perspectives.
Collapse
Affiliation(s)
- Jurjan Aman
- Department of Pulmonology, Amsterdam University Medical Center, the Netherlands (J.A.)
| | - Coert Margadant
- Department of Medical Oncology, Amsterdam University Medical Center, the NetherlandsInstitute of Biology, Leiden University, the Netherlands (C.M.)
| |
Collapse
|
2
|
Regulation of fenestra formation via actin-dynamin2 interaction in rat pituitary endothelial cells. Cell Tissue Res 2022; 390:441-451. [PMID: 36102975 DOI: 10.1007/s00441-022-03685-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 09/08/2022] [Indexed: 12/14/2022]
Abstract
Endothelial fenestrae are transcellular pores divided by a diaphragm consisting of plasmalemma vesicle-associated protein (PLVAP). They function as a channel for peptide hormones and other substances. Invagination of the plasma membrane is necessary for the fenestra formation. The actin cytoskeleton is essential for scission of endocytic vesicles from the invaginated plasma membrane. Therefore, we examined the involvement of the actin cytoskeleton in fenestra formation in cultured endothelial cells isolated from the anterior lobe (AL) of the rat pituitary, using immunofluorescence and scanning electron microscopy. Inhibition of polymerization and depolymerization of the actin cytoskeleton by latrunculin A and jasplakinolide, respectively, remarkably increased the PLVAP-positive sieve plate area and number of fenestrae. Jasplakinolide significantly affected the arrangement of the fenestra on the cell surface, resulting in parallel serpentine furrows of the fenestra. These results suggest that the actin cytoskeleton not only induces fenestra formation but also regulates cell arrangement. Dynamin is a scission protein of the invaginated plasma membrane and interacts with the actin cytoskeleton. We found that dynamin2 is mainly expressed in the endothelial cells of the rat AL. We then investigated the function of dynamin2 by the treatment with dyngo-4a, a potent inhibitor of dynamin1 and dynamin2, on the fenestra formation. As a result, the PLVAP-positive area is significantly increased by the treatment. These results show that the actin-dynamin2 interaction is essential for the control of the fenestra formation in endothelial cells of rat AL. In conclusion, the actin cytoskeleton and dynamin2 function as regulators of endothelial fenestra formation.
Collapse
|
3
|
Keum S, Yang SJ, Park E, Kang T, Choi JH, Jeong J, Hwang YE, Kim JW, Park D, Rhee S. Beta-Pix-dynamin 2 complex promotes colorectal cancer progression by facilitating membrane dynamics. Cell Oncol (Dordr) 2021; 44:1287-1305. [PMID: 34582006 PMCID: PMC8648671 DOI: 10.1007/s13402-021-00637-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Accepted: 09/14/2021] [Indexed: 12/24/2022] Open
Abstract
PURPOSE Spatiotemporal regulation of cell membrane dynamics is a major process that promotes cancer cell invasion by acting as a driving force for cell migration. Beta-Pix (βPix), a guanine nucleotide exchange factor for Rac1, has been reported to be involved in actin-mediated cellular processes, such as cell migration, by interacting with various proteins. As yet, however, the molecular mechanisms underlying βPix-mediated cancer cell invasion remain unclear. METHODS The clinical significance of βPix was analyzed in patients with colorectal cancer (CRC) using public clinical databases. Pull-down and immunoprecipitation assays were employed to identify novel binding partners for βPix. Additionally, various cell biological assays including immunocytochemistry and time-lapse video microscopy were performed to assess the effects of βPix on CRC progression. A βPix-SH3 antibody delivery system was used to determine the effects of the βPix-Dyn2 complex in CRC cells. RESULTS We found that the Src homology 3 (SH3) domain of βPix interacts with the proline-rich domain of Dynamin 2 (Dyn2), a large GTPase. The βPix-Dyn2 interaction promoted lamellipodia formation, along with plasma membrane localization of membrane-type 1 matrix metalloproteinase (MT1-MMP). Furthermore, we found that Src kinase-mediated phosphorylation of the tyrosine residue at position 442 of βPix enhanced βPix-Dyn2 complex formation. Disruption of the βPix-Dyn2 complex by βPix-SH3 antibodies targeting intracellular βPix inhibited CRC cell invasion. CONCLUSIONS Our data indicate that spatiotemporal regulation of the Src-βPix-Dyn2 axis is crucial for CRC cell invasion by promoting membrane dynamics and MT1-MMP recruitment into the leading edge. The development of inhibitors that disrupt the βPix-Dyn2 complex may be a useful therapeutic strategy for CRC.
Collapse
Affiliation(s)
- Seula Keum
- Department of Life Science, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Soo Jung Yang
- Translational Research Program, Benaroya Research Institute at Virginia Mason, Seattle, WA, 98101, USA
| | - Esther Park
- School of Life Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - TaeIn Kang
- School of Life Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Jee-Hye Choi
- Department of Life Science, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Jangho Jeong
- Department of Life Science, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Ye Eun Hwang
- Department of Life Science, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Jung-Woong Kim
- Department of Life Science, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Dongeun Park
- School of Life Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Sangmyung Rhee
- Department of Life Science, Chung-Ang University, Seoul, 06974, Republic of Korea.
| |
Collapse
|
4
|
Ricolo D, Castro-Ribera J, Araújo SJ. Cytoskeletal players in single-cell branching morphogenesis. Dev Biol 2021; 477:22-34. [PMID: 34004181 DOI: 10.1016/j.ydbio.2021.05.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 04/25/2021] [Accepted: 05/04/2021] [Indexed: 12/22/2022]
Abstract
Branching networks are a very common feature of multicellular animals and underlie the formation and function of numerous organs including the nervous system, the respiratory system, the vasculature and many internal glands. These networks range from subcellular structures such as dendritic trees to large multicellular tissues such as the lungs. The production of branched structures by single cells, so called subcellular branching, which has been better described in neurons and in cells of the respiratory and vascular systems, involves complex cytoskeletal remodelling events. In Drosophila, tracheal system terminal cells (TCs) and nervous system dendritic arborisation (da) neurons are good model systems for these subcellular branching processes. During development, the generation of subcellular branches by single-cells is characterized by extensive remodelling of the microtubule (MT) network and actin cytoskeleton, followed by vesicular transport and membrane dynamics. In this review, we describe the current knowledge on cytoskeletal regulation of subcellular branching, based on the terminal cells of the Drosophila tracheal system, but drawing parallels with dendritic branching and vertebrate vascular subcellular branching.
Collapse
Affiliation(s)
- Delia Ricolo
- Department of Genetics, Microbiology and Statistics, School of Biology, University of Barcelona, 08028, Barcelona, Spain; Institute of Biomedicine University of Barcelona (IBUB), Barcelona, Spain
| | - Judith Castro-Ribera
- Department of Genetics, Microbiology and Statistics, School of Biology, University of Barcelona, 08028, Barcelona, Spain; Institute of Biomedicine University of Barcelona (IBUB), Barcelona, Spain
| | - Sofia J Araújo
- Department of Genetics, Microbiology and Statistics, School of Biology, University of Barcelona, 08028, Barcelona, Spain; Institute of Biomedicine University of Barcelona (IBUB), Barcelona, Spain.
| |
Collapse
|
5
|
Kempers L, Wakayama Y, van der Bijl I, Furumaya C, De Cuyper IM, Jongejan A, Kat M, van Stalborch AMD, van Boxtel AL, Hubert M, Geerts D, van Buul JD, de Korte D, Herzog W, Margadant C. The endosomal RIN2/Rab5C machinery prevents VEGFR2 degradation to control gene expression and tip cell identity during angiogenesis. Angiogenesis 2021; 24:695-714. [PMID: 33983539 PMCID: PMC8292304 DOI: 10.1007/s10456-021-09788-4] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Accepted: 03/05/2021] [Indexed: 12/13/2022]
Abstract
Sprouting angiogenesis is key to many pathophysiological conditions, and is strongly regulated by vascular endothelial growth factor (VEGF) signaling through VEGF receptor 2 (VEGFR2). Here we report that the early endosomal GTPase Rab5C and its activator RIN2 prevent lysosomal routing and degradation of VEGF-bound, internalized VEGFR2 in human endothelial cells. Stabilization of endosomal VEGFR2 levels by RIN2/Rab5C is crucial for VEGF signaling through the ERK and PI3-K pathways, the expression of immediate VEGF target genes, as well as specification of angiogenic 'tip' and 'stalk' cell phenotypes and cell sprouting. Using overexpression of Rab mutants, knockdown and CRISPR/Cas9-mediated gene editing, and live-cell imaging in zebrafish, we further show that endosomal stabilization of VEGFR2 levels is required for developmental angiogenesis in vivo. In contrast, the premature degradation of internalized VEGFR2 disrupts VEGF signaling, gene expression, and tip cell formation and migration. Thus, an endosomal feedforward mechanism maintains receptor signaling by preventing lysosomal degradation, which is directly linked to the induction of target genes and cell fate in collectively migrating cells during morphogenesis.
Collapse
Affiliation(s)
- Lanette Kempers
- Sanquin Research, Plesmanlaan 125, 1066 CX, Amsterdam, The Netherlands
| | - Yuki Wakayama
- Max Planck Institute for Molecular Biomedicine, Roentgenstrasse 20, 48149, Muenster, Germany
| | - Ivo van der Bijl
- Sanquin Research, Plesmanlaan 125, 1066 CX, Amsterdam, The Netherlands
| | - Charita Furumaya
- Sanquin Research, Plesmanlaan 125, 1066 CX, Amsterdam, The Netherlands
| | - Iris M De Cuyper
- Sanquin Research, Plesmanlaan 125, 1066 CX, Amsterdam, The Netherlands
| | - Aldo Jongejan
- Department of Epidemiology and Data Science /Amsterdam Public Health Research Institute, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Marije Kat
- Sanquin Research, Plesmanlaan 125, 1066 CX, Amsterdam, The Netherlands
| | | | - Antonius L van Boxtel
- Cancer Biology and Genetics and Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA.,Developmental, Stem Cell and Cancer Biology, Swammerdam Institute for Life Sciences, University of Amsterdam, Science Park 904, 1098 XH, Amsterdam, The Netherlands
| | - Marvin Hubert
- University of Muenster, Schlossplatz 2, 48149, Muenster, Germany
| | - Dirk Geerts
- Department of Medical Biology, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Jaap D van Buul
- Sanquin Research, Plesmanlaan 125, 1066 CX, Amsterdam, The Netherlands
| | - Dirk de Korte
- Sanquin Research, Plesmanlaan 125, 1066 CX, Amsterdam, The Netherlands.,Sanquin Blood Bank, Plesmanlaan 125, 1066 CX, Amsterdam, The Netherlands
| | - Wiebke Herzog
- Max Planck Institute for Molecular Biomedicine, Roentgenstrasse 20, 48149, Muenster, Germany.,University of Muenster, Schlossplatz 2, 48149, Muenster, Germany
| | - Coert Margadant
- Angiogenesis Laboratory, Department of Medical Oncology, Amsterdam University Medical Center, location VUmc, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands.
| |
Collapse
|
6
|
Chemical Inhibitors of Dynamin Exert Differential Effects in VEGF Signaling. Cells 2021; 10:cells10050997. [PMID: 33922806 PMCID: PMC8145957 DOI: 10.3390/cells10050997] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 04/19/2021] [Accepted: 04/20/2021] [Indexed: 01/10/2023] Open
Abstract
VEGFR2 is the main receptor and mediator of the vasculogenic and angiogenic activity of VEGF. Activated VEGFR2 internalizes through clathrin-mediated endocytosis and macropinocytosis. As dynamin is a key regulator of the clathrin pathway, chemical inhibitors of dynamin are commonly used to assess the role of the clathrin route in receptor signaling. However, drugs may also exert off-target effects. Here, we compare the effects of three dynamin inhibitors, dynasore, dyngo 4a and dynole, on VEGFR2 internalization and signaling. Although these drugs consistently inhibit clathrin-mediated endocytosis of both transferrin (a typical cargo of this route) and VEGFR2, surprisingly, they exert contradictory effects in receptor signaling. Thus, while dynasore has no effect on phosphorylation of VEGFR2, the other two drugs are strong inhibitors. Furthermore, although dyngo does not interfere with phosphorylation of Akt, dynasore and dynole have a strong inhibitory effect. These inconsistent effects suggest that the above dynamin blockers, besides inhibiting dynamin-dependent endocytosis of VEGFR2, exert additional inhibitory effects on signaling that are independent of endocytosis; i.e., they are due to off-target effects. Using a recently developed protocol, we comparatively validate the specificity of two endocytic inhibitors, dynasore and EIPA. Our findings highlight the importance of assessing whether the effect of an endocytic drug on signaling is specifically due to its interference with endocytosis or due to off-targets.
Collapse
|
7
|
Märklin M, Tandler C, Kopp HG, Hoehn KL, Quintanilla-Martinez L, Borst O, Müller MR, Saur SJ. C-Cbl regulates c-MPL receptor trafficking and its internalization. J Cell Mol Med 2020; 24:12491-12503. [PMID: 32954656 PMCID: PMC7687000 DOI: 10.1111/jcmm.15785] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 07/10/2020] [Accepted: 07/31/2020] [Indexed: 01/11/2023] Open
Abstract
Thrombocyte formation from megakaryocyte and their progenitor cells is tightly regulated by thrombopoietin (TPO) and its receptor c‐MPL, thereby maintaining physiological functionality and numbers of circulating platelets. In patients, dysfunction of this regulation could cause thrombocytopenia or myeloproliferative syndromes. Since regulation of this pathway is still not completely understood, we investigated the role of the ubiquitin ligase c‐Cbl which was previously shown to negatively regulated c‐MPL signalling. We developed a new conditional mouse model using c‐Cblfl/flPf4Cre mice and demonstrated that platelet‐specific knockout of c‐Cbl led to severe microthrombocytosis and impaired uptake of TPO and c‐MPL receptor internalization. Furthermore, we characterized a constitutive STAT5 activation c‐Cbl KO platelets. This study identified c‐Cbl as a potential player in causing megakaryocytic and thrombocytic disorders.
Collapse
Affiliation(s)
- Melanie Märklin
- Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK), German Cancer Research Centre (DKFZ), University Hospital Tübingen, Tübingen, Germany
| | - Claudia Tandler
- Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK), German Cancer Research Centre (DKFZ), University Hospital Tübingen, Tübingen, Germany
| | - Hans-Georg Kopp
- Department of Molecular Oncology and Thoracic Oncology, Robert-Bosch-Hospital Stuttgart, Stuttgart, Germany
| | - Kyle L Hoehn
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW, Australia
| | | | - Oliver Borst
- Department of Kardiology and Angiology, University Hospital Tübingen, Tübingen, Germany
| | - Martin R Müller
- Department of Hematology, Oncology, Clinical Immunology and Rheumatology, University Hospital Tübingen, Tübingen, Germany.,Department of Hematology, Oncology and Immunology, Klinikum Region Hannover, KRH Klinikum Siloah, Hannover, Germany
| | - Sebastian J Saur
- Department of Hematology, Oncology, Clinical Immunology and Rheumatology, University Hospital Tübingen, Tübingen, Germany
| |
Collapse
|
8
|
Fuentes P, Sesé M, Guijarro PJ, Emperador M, Sánchez-Redondo S, Peinado H, Hümmer S, Ramón Y Cajal S. ITGB3-mediated uptake of small extracellular vesicles facilitates intercellular communication in breast cancer cells. Nat Commun 2020; 11:4261. [PMID: 32848136 PMCID: PMC7450082 DOI: 10.1038/s41467-020-18081-9] [Citation(s) in RCA: 103] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Accepted: 07/31/2020] [Indexed: 11/08/2022] Open
Abstract
Metastasis, the spread of malignant cells from a primary tumour to distant sites, causes 90% of cancer-related deaths. The integrin ITGB3 has been previously described to play an essential role in breast cancer metastasis, but the precise mechanisms remain undefined. We have now uncovered essential and thus far unknown roles of ITGB3 in vesicle uptake. The functional requirement for ITGB3 derives from its interactions with heparan sulfate proteoglycans (HSPGs) and the process of integrin endocytosis, allowing the capture of extracellular vesicles and their endocytosis-mediated internalization. Key for the function of ITGB3 is the interaction and activation of focal adhesion kinase (FAK), which is required for endocytosis of these vesicles. Thus, ITGB3 has a central role in intracellular communication via extracellular vesicles, proposed to be critical for cancer metastasis.
Collapse
Affiliation(s)
- Pedro Fuentes
- Translational Molecular Pathology, Vall d'Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona (UAB), Barcelona, Spain
- Spanish Biomedical Research Network Centre in Oncology (CIBERONC), Madrid, Spain
| | - Marta Sesé
- Translational Molecular Pathology, Vall d'Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona (UAB), Barcelona, Spain
- Spanish Biomedical Research Network Centre in Oncology (CIBERONC), Madrid, Spain
| | - Pedro J Guijarro
- Translational Molecular Pathology, Vall d'Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona (UAB), Barcelona, Spain
| | - Marta Emperador
- Translational Molecular Pathology, Vall d'Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona (UAB), Barcelona, Spain
- Tumor Biomarkers Group, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Sara Sánchez-Redondo
- Microenvironment & Metastasis Group, Molecular Oncology Program, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Héctor Peinado
- Microenvironment & Metastasis Group, Molecular Oncology Program, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Stefan Hümmer
- Translational Molecular Pathology, Vall d'Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona (UAB), Barcelona, Spain.
- Spanish Biomedical Research Network Centre in Oncology (CIBERONC), Madrid, Spain.
| | - Santiago Ramón Y Cajal
- Translational Molecular Pathology, Vall d'Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona (UAB), Barcelona, Spain.
- Spanish Biomedical Research Network Centre in Oncology (CIBERONC), Madrid, Spain.
| |
Collapse
|
9
|
Mousavi SA, Skjeldal F, Fønhus MS, Haugen LH, Eskild W, Berg T, Bakke O. Receptor-Mediated Endocytosis of VEGF-A in Rat Liver Sinusoidal Endothelial Cells. BIOMED RESEARCH INTERNATIONAL 2019; 2019:5496197. [PMID: 31583245 PMCID: PMC6754870 DOI: 10.1155/2019/5496197] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 06/21/2019] [Accepted: 07/02/2019] [Indexed: 12/25/2022]
Abstract
BACKGROUND AND AIMS Vascular endothelial growth factor (VEGF) receptors (VEGFR1 and VEGFR2) bind VEGF-A with high affinity. This study sought to determine the relative contributions of these two receptors to receptor-mediated endocytosis of VEGF-A and to clarify their endocytic itineraries in rat liver sinusoidal endothelial cells (LSECs). METHODS Isolated LSECs and radiolabeled VEGF-A were used to examine surface binding and receptor-mediated endocytosis. Quantitative real time RT-PCR (Q-RT-PCR) and Western blotting were applied to demonstrate receptor expression. RESULTS Q-RT-PCR analysis showed that VEGFR1 and VEGFR2 mRNA were expressed in LSECs. Ligand saturation analysis at 4°C indicated two different classes of [125I]-VEGFA binding sites on LSECs with apparent dissociation constants of 8 and 210 pM. At 37°C, LSECs efficiently took up and degraded [125I]-VEGF-A for at least 2 hours. Uptake of [125I]-VEGF-A by LSECs was blocked by dynasore that inhibits dynamin-dependent internalization, whereas inhibition of cysteine proteases by leupeptin inhibited degradation without affecting the uptake of [125I]-VEGF-A, suggesting that it is degraded following transport to lysosomes. Incubation of LSECs in the continued presence of a saturating concentration of unlabeled VEGF-A at 37°C was associated with a loss of as much as 75% of the total VEGFR2 within 30 min as shown by Western blot analysis, whereas there was no appreciable decrease in protein levels for VEGFR1 after 120 min incubation, suggesting that VEGF-A stimulation downregulates VEGFR2, but not VEGFR1, in LSECs. This possibility was supported by the observation that a hexapeptide that specifically blocks VEGF-A binding to VEGFR1 caused a marked reduction in the uptake of [125I]-VEGF-A, whereas a control peptide had no effect. Finally, live cell imaging studies using a fluorescently labeled anti-VEGFR2 antibody showed that VEGFR2 was transported via early and late endosomes to reach endolysosomes where degradation of the VEGFR2 takes place. CONCLUSION Our studies suggest that, subsequent to VEGF-A binding and internalization, the unoccupied VEGFR1 may recycle to the cell surface allowing its reutilization, whereas the majority of the internalized VEGFR2 is targeted for degradation.
Collapse
Affiliation(s)
- Seyed Ali Mousavi
- Department of Immunology and Transfusion Medicine, Akershus University Hospital, University of Oslo, Norway
- Department of Biosciences, University of Oslo, Norway
| | | | | | | | - Winnie Eskild
- Department of Biosciences, University of Oslo, Norway
| | - Trond Berg
- Department of Biosciences, University of Oslo, Norway
| | - Oddmund Bakke
- Department of Biosciences, University of Oslo, Norway
| |
Collapse
|
10
|
Peterson SM, Turner JE, Harrington A, Davis-Knowlton J, Lindner V, Gridley T, Vary CPH, Liaw L. Notch2 and Proteomic Signatures in Mouse Neointimal Lesion Formation. Arterioscler Thromb Vasc Biol 2018; 38:1576-1593. [PMID: 29853569 PMCID: PMC6023756 DOI: 10.1161/atvbaha.118.311092] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2017] [Accepted: 05/16/2018] [Indexed: 12/29/2022]
Abstract
Supplemental Digital Content is available in the text. Objective— Vascular remodeling is associated with complex molecular changes, including increased Notch2, which promotes quiescence in human smooth muscle cells. We used unbiased protein profiling to understand molecular signatures related to neointimal lesion formation in the presence or absence of Notch2 and to test the hypothesis that loss of Notch2 would increase neointimal lesion formation because of a hyperproliferative injury response. Approach and Results— Murine carotid arteries isolated at 6 or 14 days after ligation injury were analyzed by mass spectrometry using a data-independent acquisition strategy in comparison to uninjured or sham injured arteries. We used a tamoxifen-inducible, cell-specific Cre recombinase strain to delete the Notch2 gene in smooth muscle cells. Vessel morphometric analysis and immunohistochemical staining were used to characterize lesion formation, assess vascular smooth muscle cell proliferation, and validate proteomic findings. Loss of Notch2 in smooth muscle cells leads to protein profile changes in the vessel wall during remodeling but does not alter overall lesion morphology or cell proliferation. Loss of smooth muscle Notch2 also decreases the expression of enhancer of rudimentary homolog, plectin, and annexin A2 in vascular remodeling. Conclusions— We identified unique protein signatures that represent temporal changes in the vessel wall during neointimal lesion formation in the presence and absence of Notch2. Overall lesion formation was not affected with loss of smooth muscle Notch2, suggesting compensatory pathways. We also validated the regulation of known injury- or Notch-related targets identified in other vascular contexts, providing additional insight into conserved pathways involved in vascular remodeling.
Collapse
Affiliation(s)
- Sarah M Peterson
- From the Maine Medical Center Research Institute, Scarborough (S.M.P., J.E.T., A.H., J.D.-K., V.L., T.G., C.P.H.V., L.L.).,University of Maine Graduate School of Biomedical Science and Engineering, Orono (S.M.P., V.L., T.G., C.P.H.V., L.L.)
| | - Jacqueline E Turner
- From the Maine Medical Center Research Institute, Scarborough (S.M.P., J.E.T., A.H., J.D.-K., V.L., T.G., C.P.H.V., L.L.)
| | - Anne Harrington
- From the Maine Medical Center Research Institute, Scarborough (S.M.P., J.E.T., A.H., J.D.-K., V.L., T.G., C.P.H.V., L.L.)
| | - Jessica Davis-Knowlton
- From the Maine Medical Center Research Institute, Scarborough (S.M.P., J.E.T., A.H., J.D.-K., V.L., T.G., C.P.H.V., L.L.).,Tufts Sackler School of Graduate Biomedical Sciences, Boston, MA (J.D.-K., V.L., T.G., C.P.H.V., L.L.)
| | - Volkhard Lindner
- From the Maine Medical Center Research Institute, Scarborough (S.M.P., J.E.T., A.H., J.D.-K., V.L., T.G., C.P.H.V., L.L.).,University of Maine Graduate School of Biomedical Science and Engineering, Orono (S.M.P., V.L., T.G., C.P.H.V., L.L.).,Tufts Sackler School of Graduate Biomedical Sciences, Boston, MA (J.D.-K., V.L., T.G., C.P.H.V., L.L.)
| | - Thomas Gridley
- From the Maine Medical Center Research Institute, Scarborough (S.M.P., J.E.T., A.H., J.D.-K., V.L., T.G., C.P.H.V., L.L.).,University of Maine Graduate School of Biomedical Science and Engineering, Orono (S.M.P., V.L., T.G., C.P.H.V., L.L.).,Tufts Sackler School of Graduate Biomedical Sciences, Boston, MA (J.D.-K., V.L., T.G., C.P.H.V., L.L.)
| | - Calvin P H Vary
- From the Maine Medical Center Research Institute, Scarborough (S.M.P., J.E.T., A.H., J.D.-K., V.L., T.G., C.P.H.V., L.L.).,University of Maine Graduate School of Biomedical Science and Engineering, Orono (S.M.P., V.L., T.G., C.P.H.V., L.L.).,Tufts Sackler School of Graduate Biomedical Sciences, Boston, MA (J.D.-K., V.L., T.G., C.P.H.V., L.L.)
| | - Lucy Liaw
- From the Maine Medical Center Research Institute, Scarborough (S.M.P., J.E.T., A.H., J.D.-K., V.L., T.G., C.P.H.V., L.L.) .,University of Maine Graduate School of Biomedical Science and Engineering, Orono (S.M.P., V.L., T.G., C.P.H.V., L.L.).,Tufts Sackler School of Graduate Biomedical Sciences, Boston, MA (J.D.-K., V.L., T.G., C.P.H.V., L.L.)
| |
Collapse
|
11
|
Yao Z, Wang X, Zhang W, Liu Y, Ni T. Photochemical tissue bonding promotes the proliferation and migration of injured tenocytes through ROS/RhoA/NF-κB/Dynamin 2 signaling pathway. J Cell Physiol 2018; 233:7047-7056. [PMID: 29744878 DOI: 10.1002/jcp.26628] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Accepted: 03/30/2018] [Indexed: 12/26/2022]
Abstract
Photochemical tissue bonding (PTB) has been found to promote the healing of Achilles tendon tissue injury and to reduce postoperative complications. However, the underlying cellular and molecular mechanisms are not clear. In this study, the cell proliferation, ROS generation, migration and the protein expression of DNM2, NF-κB p65, TGF-β1 and VEGF in tenocytes after PTB treatment were measured by CCK-8, flow cytometry, Transwell and western blot assay, respectively. And those in tenocytes after DNM2 silencing or overexpressing or treatment with inhibitors of NF-κB, ROS and RhoA were also measured. Our results showed that 10 mW PTB treatment for 80 and 120 s significantly increased cell proliferation and increased ROS generation in tenocytes. 10 mW PTB treatment for 40 and 80 s significantly activated RhoA and increased the protein expression of DNM2, NF-κB p65, TGF-β1 and VEGF, but 10 mW PTB treatment for 120 s decreased the protein expression of those. DNM2 silencing significantly suppressed cell migration and the expression of DNM2, TGF-β1, and VEGF in tenocytes after PTB treatment (10 mW, 80 s), which was inhibited by DNM2 overexpression. Individual treatment with inhibitor of NF-κB, ROS, and RhoA in tenocytes showed decreased protein expression of DNM2, TGF-β1, and VEGF. Moreover, in vivo experiment found that PTB treatment significantly inhibited cell apoptosis and the expression of DNM2, NF-κB p65, RhoA, TGF-β1, and VEGF in a time-dependent manner. Taken together, our results suggest that PTB promotes the proliferation and migration of injured tenocytes through ROS/RhoA/NF-κB/DNM2 signaling pathway.
Collapse
Affiliation(s)
- Zuochao Yao
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People' Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xi Wang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People' Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Wen Zhang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People' Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yushu Liu
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People' Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Tao Ni
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People' Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
12
|
Reventun P, Alique M, Cuadrado I, Márquez S, Toro R, Zaragoza C, Saura M. iNOS-Derived Nitric Oxide Induces Integrin-Linked Kinase Endocytic Lysosome-Mediated Degradation in the Vascular Endothelium. Arterioscler Thromb Vasc Biol 2017; 37:1272-1281. [PMID: 28546219 DOI: 10.1161/atvbaha.117.309560] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Accepted: 05/08/2017] [Indexed: 12/22/2022]
Abstract
Objective—
ILK (integrin-linked kinase) plays a key role in controlling vasomotor tone and is decreased in atherosclerosis. The objective of this study is to test whether nitric oxide (NO) regulates ILK in vascular remodeling.
Approach and Results—
We found a striking correlation between increased levels of inducible nitric oxide and decreased ILK levels in human atherosclerosis and in a mouse model of vascular remodeling (carotid artery ligation) comparing with iNOS (inducible NO synthase) knockout mice. iNOS induction produced the same result in mouse aortic endothelial cells, and these effects were mimicked by an NO donor in a time-dependent manner. We found that NO decreased ILK protein stability by promoting the dissociation of the complex ILK/Hsp90 (heat shock protein 90)/eNOS (endothelial NO synthase), leading to eNOS uncoupling. NO also destabilized ILK signaling platform and lead to decreased levels of paxillin and α-parvin. ILK phosphorylation of its downstream target GSK3-β (glycogen synthase kinase 3 beta) was decreased by NO. Mechanistically, NO increased ILK ubiquitination mediated by the E3 ubiquitin ligase CHIP (C terminus of HSC70-interacting protein), but ILK ubiquitination was not followed by proteasome degradation. Alternatively, NO drove ILK to degradation through the endocytic-lysosomal pathway. ILK colocalized with the lysosome marker LAMP-1 (lysosomal-associated membrane protein 1) in endothelial cells, and inhibition of lysosome activity with chloroquine reversed the effect of NO. Likewise, ILK colocalized with the early endosome marker EEA1 (early endosome antigen 1). ILK endocytosis proceeded via dynamin because a specific inhibitor of dynamin (Dyngo 4a) was able to reverse ILK endocytosis and its lysosome degradation.
Conclusions—
Endocytosis regulates ILK signaling in vascular remodeling where there is an overload of inducible NO, and thus its inhibition may represent a novel target to fight atherosclerotic disease.
Collapse
Affiliation(s)
- Paula Reventun
- From the Biology Systems Department, Physiology, School of Medicine and Health Sciences, Universidad Alcalá (IRYCIS), Madrid, Spain (P.R., M.A., S.M., M.S.); Cardiology Department, University Francisco de Vitoria/Hospital Ramón y Cajal Research Unit (IRYCIS), Madrid, Spain (C.Z.); and Cardiology Department, School of Medicine, Cádiz University, Spain (R.T.)
| | - Matilde Alique
- From the Biology Systems Department, Physiology, School of Medicine and Health Sciences, Universidad Alcalá (IRYCIS), Madrid, Spain (P.R., M.A., S.M., M.S.); Cardiology Department, University Francisco de Vitoria/Hospital Ramón y Cajal Research Unit (IRYCIS), Madrid, Spain (C.Z.); and Cardiology Department, School of Medicine, Cádiz University, Spain (R.T.)
| | - Irene Cuadrado
- From the Biology Systems Department, Physiology, School of Medicine and Health Sciences, Universidad Alcalá (IRYCIS), Madrid, Spain (P.R., M.A., S.M., M.S.); Cardiology Department, University Francisco de Vitoria/Hospital Ramón y Cajal Research Unit (IRYCIS), Madrid, Spain (C.Z.); and Cardiology Department, School of Medicine, Cádiz University, Spain (R.T.)
| | - Susana Márquez
- From the Biology Systems Department, Physiology, School of Medicine and Health Sciences, Universidad Alcalá (IRYCIS), Madrid, Spain (P.R., M.A., S.M., M.S.); Cardiology Department, University Francisco de Vitoria/Hospital Ramón y Cajal Research Unit (IRYCIS), Madrid, Spain (C.Z.); and Cardiology Department, School of Medicine, Cádiz University, Spain (R.T.)
| | - Rocío Toro
- From the Biology Systems Department, Physiology, School of Medicine and Health Sciences, Universidad Alcalá (IRYCIS), Madrid, Spain (P.R., M.A., S.M., M.S.); Cardiology Department, University Francisco de Vitoria/Hospital Ramón y Cajal Research Unit (IRYCIS), Madrid, Spain (C.Z.); and Cardiology Department, School of Medicine, Cádiz University, Spain (R.T.)
| | - Carlos Zaragoza
- From the Biology Systems Department, Physiology, School of Medicine and Health Sciences, Universidad Alcalá (IRYCIS), Madrid, Spain (P.R., M.A., S.M., M.S.); Cardiology Department, University Francisco de Vitoria/Hospital Ramón y Cajal Research Unit (IRYCIS), Madrid, Spain (C.Z.); and Cardiology Department, School of Medicine, Cádiz University, Spain (R.T.)
| | - Marta Saura
- From the Biology Systems Department, Physiology, School of Medicine and Health Sciences, Universidad Alcalá (IRYCIS), Madrid, Spain (P.R., M.A., S.M., M.S.); Cardiology Department, University Francisco de Vitoria/Hospital Ramón y Cajal Research Unit (IRYCIS), Madrid, Spain (C.Z.); and Cardiology Department, School of Medicine, Cádiz University, Spain (R.T.)
| |
Collapse
|
13
|
Basagiannis D, Zografou S, Galanopoulou K, Christoforidis S. Dynasore impairs VEGFR2 signalling in an endocytosis-independent manner. Sci Rep 2017; 7:45035. [PMID: 28327657 PMCID: PMC5361198 DOI: 10.1038/srep45035] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Accepted: 02/17/2017] [Indexed: 12/17/2022] Open
Abstract
VEGFR2 is a critical angiogenic receptor playing a key role in vascular homeostasis. Upon activation by VEGF, VEGFR2 becomes endocytosed. Internalisation of VEGFR2 is facilitated, in part, through clathrin mediated endocytosis (CME), the role of which in VEGFR2 function is debated. Here, we confirm the contribution of CME in VEGFR2 uptake. However, curiously, we find that different approaches of inhibition of CME exert contradictory effects on VEGF signalling; knockdown of clathrin, or of dynamin, or overexpression of dynamin K44A, do not affect VEGF-induced phosphorylation of ERK1/2, while dynasore causes strong inhibition. We resolve this discrepancy by showing that although dynasore inhibits CME of VEGFR2, its inhibitory action in ERK1/2 phosphorylation is not related to attenuation of VEGFR2 endocytosis; it is rather due to an off-target effect of the drug. Dynasore inhibits VEGF-induced calcium release, a signalling event that lies upstream of ERK1/2, which implies that this effect could be responsible, at least in part, for the inhibitory action of the drug on VEGF-to-ERK1/2 signalling. These results raise caution that although dynasore is specific in inhibiting clathrin- and dynamin-mediated endocytosis, it may also exert off-target effects on signalling molecules, hence influencing the interpretation of the role of endocytosis in signalling.
Collapse
Affiliation(s)
- Dimitris Basagiannis
- Institute of Molecular Biology and Biotechnology-Biomedical Research, Foundation for Research and Technology, 45110 Ioannina, Greece.,Laboratory of Biological Chemistry, Department of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece
| | - Sofia Zografou
- Institute of Molecular Biology and Biotechnology-Biomedical Research, Foundation for Research and Technology, 45110 Ioannina, Greece
| | - Katerina Galanopoulou
- Institute of Molecular Biology and Biotechnology-Biomedical Research, Foundation for Research and Technology, 45110 Ioannina, Greece.,Laboratory of Biological Chemistry, Department of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece
| | - Savvas Christoforidis
- Institute of Molecular Biology and Biotechnology-Biomedical Research, Foundation for Research and Technology, 45110 Ioannina, Greece.,Laboratory of Biological Chemistry, Department of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece
| |
Collapse
|
14
|
Eppler FJ, Quast T, Kolanus W. Dynamin2 controls Rap1 activation and integrin clustering in human T lymphocyte adhesion. PLoS One 2017; 12:e0172443. [PMID: 28273099 PMCID: PMC5342215 DOI: 10.1371/journal.pone.0172443] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Accepted: 01/23/2017] [Indexed: 11/19/2022] Open
Abstract
Leukocyte trafficking is crucial to facilitate efficient immune responses. Here, we report that the large GTPase dynamin2, which is generally considered to have a key role in endocytosis and membrane remodeling, is an essential regulator of integrin-dependent human T lymphocyte adhesion and migration. Chemical inhibition or knockdown of dynamin2 expression significantly reduced integrin-dependent T cell adhesion in vitro. This phenotype was not observed when T cells were treated with various chemical inhibitors which abrogate endocytosis or actin polymerization. We furthermore detected dynamin2 in signaling complexes and propose that it controls T cell adhesion via FAK/Pyk2- and RapGEF1-mediated Rap1 activation. In addition, the dynamin2 inhibitor-induced reduction of lymphocyte adhesion can be rescued by Rap1a overexpression. We demonstrate that the dynamin2 effect on T cell adhesion does not involve integrin affinity regulation but instead relies on its ability to modulate integrin valency. Taken together, we suggest a previously unidentified role of dynamin2 in the regulation of integrin-mediated lymphocyte adhesion via a Rap1 signaling pathway.
Collapse
Affiliation(s)
- Felix J. Eppler
- Division of Molecular Immunology and Cell Biology, Life and Medical Sciences Institute (LIMES), University of Bonn, Bonn, Germany
| | - Thomas Quast
- Division of Molecular Immunology and Cell Biology, Life and Medical Sciences Institute (LIMES), University of Bonn, Bonn, Germany
| | - Waldemar Kolanus
- Division of Molecular Immunology and Cell Biology, Life and Medical Sciences Institute (LIMES), University of Bonn, Bonn, Germany
- * E-mail:
| |
Collapse
|
15
|
Évora A, de Freitas V, Mateus N, Fernandes I. The effect of anthocyanins from red wine and blackberry on the integrity of a keratinocyte model using ECIS. Food Funct 2017; 8:3989-3998. [DOI: 10.1039/c7fo01239j] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Anthocyanins enhanced the healing rate of keratinocyte cells.
Collapse
Affiliation(s)
- Ana Évora
- REQUIMTE/LAQV
- Department of Chemistry and Biochemistry
- Faculty of Sciences
- University of Porto
- Porto
| | - Victor de Freitas
- REQUIMTE/LAQV
- Department of Chemistry and Biochemistry
- Faculty of Sciences
- University of Porto
- Porto
| | - Nuno Mateus
- REQUIMTE/LAQV
- Department of Chemistry and Biochemistry
- Faculty of Sciences
- University of Porto
- Porto
| | - Iva Fernandes
- REQUIMTE/LAQV
- Department of Chemistry and Biochemistry
- Faculty of Sciences
- University of Porto
- Porto
| |
Collapse
|
16
|
Wang R, Ding Q, De Assuncao TM, Mounajjed T, Maiers JL, Dou C, Cao S, Yaqoob U, Huebert RC, Shah VH. Hepatic Stellate Cell Selective Disruption of Dynamin-2 GTPase Increases Murine Fibrogenesis through Up-Regulation of Sphingosine-1 Phosphate-Induced Cell Migration. THE AMERICAN JOURNAL OF PATHOLOGY 2016; 187:134-145. [PMID: 27840081 PMCID: PMC5225297 DOI: 10.1016/j.ajpath.2016.09.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Revised: 08/22/2016] [Accepted: 09/01/2016] [Indexed: 01/04/2023]
Abstract
Dynamin-2 (Dyn2) is implicated in endocytosis of receptor tyrosine kinases, which contribute to hepatic stellate cell (HSC) activation and liver fibrosis. A point mutation converting lysine 44 of Dyn2 to alanine (Dyn2K44A) disrupts its GTPase activity. We hypothesized that Dyn2K44A expression in HSCs would decrease HSC activation and fibrogenesis in vivo by disrupting receptor tyrosine kinase endocytosis and signaling. Dyn2K44Afl/fl mice were crossed with Collagen1-Cre (Col1Cre) mice to generate offspring with HSC selective expression of Dyn2K44A (Col1Cre/Dyn2K44Afl/fl). Contrary to our hypothesis, Col1Cre/Dyn2K44Afl/fl mice showed increased hepatic fibrosis in response to liver injury. To elucidate mechanisms, we conducted in vitro experiments with HSCs infected with adenoviral vectors encoding LacZ, Dyn2K44A, or Dyn2WT. HSC-expressing Dyn2K44A displayed increased mRNA and protein levels of sphingosine kinase-1 (SK1), an enzyme previously implicated in the pathogenesis of fibrosis. To study the functional effects of Dyn2K44A regulation of SK1, we examined effects of AKT signaling and migration in HSCs. Dyn2K44A promoted both AKT phosphorylation and HSC migration in an SK1-dependent manner. Genetic disruption of Dyn2 GTPase activity selectively in HSC enhances fibrogenesis, driven at least in part through up-regulation of the SK1 pathway and cell migration in HSCs.
Collapse
Affiliation(s)
- Ruisi Wang
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota
| | - Qian Ding
- Department of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Thiago M De Assuncao
- Department of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Taofic Mounajjed
- Laboratory of Anatomic Pathology, Mayo Clinic, Rochester, Minnesota
| | - Jessica L Maiers
- Department of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Changwei Dou
- Department of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Sheng Cao
- Department of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Usman Yaqoob
- Department of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Robert C Huebert
- Department of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Vijay H Shah
- Department of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota.
| |
Collapse
|
17
|
Chalick M, Jacobi O, Pichinuk E, Garbar C, Bensussan A, Meeker A, Ziv R, Zehavi T, Smorodinsky NI, Hilkens J, Hanisch FG, Rubinstein DB, Wreschner DH. MUC1-ARF-A Novel MUC1 Protein That Resides in the Nucleus and Is Expressed by Alternate Reading Frame Translation of MUC1 mRNA. PLoS One 2016; 11:e0165031. [PMID: 27768738 PMCID: PMC5074479 DOI: 10.1371/journal.pone.0165031] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2015] [Accepted: 10/05/2016] [Indexed: 01/26/2023] Open
Abstract
Translation of mRNA in alternate reading frames (ARF) is a naturally occurring process heretofore underappreciated as a generator of protein diversity. The MUC1 gene encodes MUC1-TM, a signal-transducing trans-membrane protein highly expressed in human malignancies. Here we show that an AUG codon downstream to the MUC1-TM initiation codon initiates an alternate reading frame thereby generating a novel protein, MUC1-ARF. MUC1-ARF, like its MUC1-TM 'parent’ protein, contains a tandem repeat (VNTR) domain. However, the amino acid sequence of the MUC1-ARF tandem repeat as well as N- and C- sequences flanking it differ entirely from those of MUC1-TM. In vitro protein synthesis assays and extensive immunohistochemical as well as western blot analyses with MUC1-ARF specific monoclonal antibodies confirmed MUC1-ARF expression. Rather than being expressed at the cell membrane like MUC1-TM, immunostaining showed that MUC1-ARF protein localizes mainly in the nucleus: Immunohistochemical analyses of MUC1-expressing tissues demonstrated MUC1-ARF expression in the nuclei of secretory luminal epithelial cells. MUC1-ARF expression varies in different malignancies. While the malignant epithelial cells of pancreatic cancer show limited expression, in breast cancer tissue MUC1-ARF demonstrates strong nuclear expression. Proinflammatory cytokines upregulate expression of MUC1-ARF protein and co-immunoprecipitation analyses demonstrate association of MUC1-ARF with SH3 domain-containing proteins. Mass spectrometry performed on proteins coprecipitating with MUC1-ARF demonstrated Glucose-6-phosphate 1-dehydrogenase (G6PD) and Dynamin 2 (DNM2). These studies not only reveal that the MUC1 gene generates a previously unidentified MUC1-ARF protein, they also show that just like its ‘parent’ MUC1-TM protein, MUC1-ARF is apparently linked to signaling and malignancy, yet a definitive link to these processes and the roles it plays awaits a precise identification of its molecular functions. Comprising at least 524 amino acids, MUC1-ARF is, furthermore, the longest ARF protein heretofore described.
Collapse
Affiliation(s)
- Michael Chalick
- Department of Cell Research and Immunology, Tel Aviv University, Ramat Aviv, Israel
| | - Oded Jacobi
- Department of Cell Research and Immunology, Tel Aviv University, Ramat Aviv, Israel
| | - Edward Pichinuk
- Department of Cell Research and Immunology, Tel Aviv University, Ramat Aviv, Israel
| | - Christian Garbar
- Department of Biopathology, Institut Jean-Godinot, Reims Cedex, France
| | | | - Alan Meeker
- The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Ravit Ziv
- Department of Cell Research and Immunology, Tel Aviv University, Ramat Aviv, Israel
| | - Tania Zehavi
- Department of Pathology, Meir Medical Center, Kfar Saba, Israel
| | | | - John Hilkens
- Division of Molecular Genetics, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Franz-Georg Hanisch
- Institute of Biochemistry II, Medical Faculty, University of Cologne, Köln, Germany
| | | | - Daniel H. Wreschner
- Department of Cell Research and Immunology, Tel Aviv University, Ramat Aviv, Israel
- * E-mail:
| |
Collapse
|
18
|
Basagiannis D, Zografou S, Murphy C, Fotsis T, Morbidelli L, Ziche M, Bleck C, Mercer J, Christoforidis S. VEGF induces signalling and angiogenesis by directing VEGFR2 internalisation through macropinocytosis. J Cell Sci 2016; 129:4091-4104. [PMID: 27656109 DOI: 10.1242/jcs.188219] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Accepted: 09/13/2016] [Indexed: 01/15/2023] Open
Abstract
Endocytosis plays a crucial role in receptor signalling. VEGFR2 (also known as KDR) and its ligand VEGFA are fundamental in neovascularisation. However, our understanding of the role of endocytosis in VEGFR2 signalling remains limited. Despite the existence of diverse internalisation routes, the only known endocytic pathway for VEGFR2 is the clathrin-mediated pathway. Here, we show that this pathway is the predominant internalisation route for VEGFR2 only in the absence of ligand. Intriguingly, VEGFA induces a new internalisation itinerary for VEGFR2, the pathway of macropinocytosis, which becomes the prevalent endocytic route for the receptor in the presence of ligand, whereas the contribution of the clathrin-mediated route becomes minor. Macropinocytic internalisation of VEGFR2, which mechanistically is mediated through the small GTPase CDC42, takes place through macropinosomes generated at ruffling areas of the membrane. Interestingly, macropinocytosis plays a crucial role in VEGFA-induced signalling, endothelial cell functions in vitro and angiogenesis in vivo, whereas clathrin-mediated endocytosis is not essential for VEGFA signalling. These findings expand our knowledge on the endocytic pathways of VEGFR2 and suggest that VEGFA-driven internalisation of VEGFR2 through macropinocytosis is essential for endothelial cell signalling and angiogenesis.
Collapse
Affiliation(s)
- Dimitris Basagiannis
- Institute of Molecular Biology and Biotechnology-Biomedical Research, Foundation for Research and Technology, Ioannina 45110, Greece.,Laboratory of Biological Chemistry, Department of Medicine, School of Health Sciences, University of Ioannina, Ioannina 45110, Greece
| | - Sofia Zografou
- Institute of Molecular Biology and Biotechnology-Biomedical Research, Foundation for Research and Technology, Ioannina 45110, Greece
| | - Carol Murphy
- Institute of Molecular Biology and Biotechnology-Biomedical Research, Foundation for Research and Technology, Ioannina 45110, Greece.,School of Biosciences, College of Life and Environmental Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| | - Theodore Fotsis
- Institute of Molecular Biology and Biotechnology-Biomedical Research, Foundation for Research and Technology, Ioannina 45110, Greece.,Laboratory of Biological Chemistry, Department of Medicine, School of Health Sciences, University of Ioannina, Ioannina 45110, Greece
| | - Lucia Morbidelli
- Department of Life Sciences, University of Siena, Via Aldo Moro 2, Siena 53100, Italy
| | - Marina Ziche
- Department of Life Sciences, University of Siena, Via Aldo Moro 2, Siena 53100, Italy
| | | | - Jason Mercer
- Institute of Biochemistry, ETH, Zurich 8093, Switzerland.,MRC-Laboratory for Molecular Cell Biology, University College London, Gower Street, London WC1E 6BT, UK
| | - Savvas Christoforidis
- Institute of Molecular Biology and Biotechnology-Biomedical Research, Foundation for Research and Technology, Ioannina 45110, Greece .,Laboratory of Biological Chemistry, Department of Medicine, School of Health Sciences, University of Ioannina, Ioannina 45110, Greece
| |
Collapse
|
19
|
Basagiannis D, Christoforidis S. Constitutive Endocytosis of VEGFR2 Protects the Receptor against Shedding. J Biol Chem 2016; 291:16892-903. [PMID: 27298320 DOI: 10.1074/jbc.m116.730309] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Indexed: 12/19/2022] Open
Abstract
VEGFR2 plays a fundamental role in blood vessel formation and in life threatening diseases, such as cancer angiogenesis and cardiovascular disorders. Although inactive growth factor receptors are mainly localized at the plasma membrane, VEGFR2 undergoes constitutive endocytosis (in the absence of ligand) and recycling. Intriguingly, the significance of these futile transport cycles of VEGFR2 remains unclear. Here we found that, unexpectedly, the function of constitutive endocytosis of VEGFR2 is to protect the receptor against plasma membrane cleavage (shedding), thereby preserving the functional state of the receptor until the time of activation by VEGF. Inhibition of constitutive endocytosis of VEGFR2, by interference with the function of clathrin, dynamin, or Rab5, increases dramatically the cleavage/shedding of VEGFR2. Shedding of VEGFR2 produces an N-terminal soluble fragment (100 kDa, s100), which is released in the extracellular space, and a residual C-terminal part (130 kDa, p130) that remains integrated at the plasma membrane. The released soluble fragment (s100) co-immunoprecipitates with VEGF, in line with the topology of the VEGF-binding domain at the N terminus of VEGFR2. Increased shedding of VEGFR2 (via inhibition of constitutive endocytosis) results in reduced response to VEGF, consistently with the loss of the VEGF-binding domain from the membrane remnant of VEGFR2. These data suggest that constitutive internalization of VEGFR2 protects the receptor against shedding and provides evidence for an unprecedented mechanism via which endocytosis can regulate the fate and activity of growth factor receptors.
Collapse
Affiliation(s)
- Dimitris Basagiannis
- From the Institute of Molecular Biology and Biotechnology-Biomedical Research, Foundation for Research and Technology, 45110 Ioannina and the Department of Medicine, Laboratory of Biological Chemistry, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece
| | - Savvas Christoforidis
- From the Institute of Molecular Biology and Biotechnology-Biomedical Research, Foundation for Research and Technology, 45110 Ioannina and the Department of Medicine, Laboratory of Biological Chemistry, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece
| |
Collapse
|
20
|
Tsou JH, Yang YC, Pao PC, Lin HC, Huang NK, Lin ST, Hsu KS, Yeh CM, Lee KH, Kuo CJ, Yang DM, Lin JH, Chang WC, Lee YC. Important Roles of Ring Finger Protein 112 in Embryonic Vascular Development and Brain Functions. Mol Neurobiol 2016; 54:2286-2300. [PMID: 26951452 DOI: 10.1007/s12035-016-9812-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Accepted: 02/22/2016] [Indexed: 11/28/2022]
Abstract
Rnf112 is a member of the RING finger protein family. The expression of Rnf112 is abundant in the brain and is regulated during brain development. Our previous study has revealed that Rnf112 can promote neuronal differentiation by inhibiting the progression of the cell cycle in cell models. In this study, we further revealed the important functions of Rnf112 in embryo development and in adult brain. Our data showed that most of the Rnf112 -/- embryos exhibited blood vascular defects and died in utero. Upon further investigation, we found that the survival rate of homozygous Rnf112 knockout mice in 129/sv and C57BL/6 mixed genetic background was increased. The survived newborns of Rnf112 -/- mice manifested growth retardation as indicated by smaller size and a reduced weight. Although the overall organization of the brain did not appear to be severely affected in Rnf112 -/- mice, using in vivo 3D MRI imaging, we found that when compared to wild-type littermates, brains of Rnf112 -/- mice were smaller. In addition, Rnf112 -/- mice displayed impairment of brain functions including motor balance, and spatial learning and memory. Our results provide important aspects for the study of Rnf112 gene functions.
Collapse
Affiliation(s)
- Jen-Hui Tsou
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Ying-Chen Yang
- Department of Biotechnology and Animal Science, College of Bioresources, National Ilan University, Ilan, Taiwan
| | - Ping-Chieh Pao
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Hui-Ching Lin
- Department and Institute of Physiology, School of Medicine, National Yang-Ming University, Taipei, Taiwan.,Brain Research Center, National Yang-Ming University, Taipei, Taiwan
| | - Nai-Kuei Huang
- National Research Institute of Chinese Medicine, Taipei, Taiwan.,Ph.D. Program for Neural Regenerative Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei, 11031, Taiwan
| | - Shih-Ting Lin
- Department of Pharmacology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Kuei-Sen Hsu
- Department of Pharmacology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Che-Ming Yeh
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Kuen-Haur Lee
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Chu-Jen Kuo
- Ph.D. Program for Neural Regenerative Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei, 11031, Taiwan.,Department of Radiology, Shin Kong Wu Ho-Su Memorial Hospital, School of Medicine, Fu Jen Catholic University, Taipei, Taiwan
| | - De-Ming Yang
- Department of Medical Research and Education, Taipei Veterans General Hospital, Taipei, Taiwan.,Institute of Biophotonics, School of Medical Technology and Engineering, National Yang-Ming University, Taipei, Taiwan
| | - Jiann-Her Lin
- Ph.D. Program for Neural Regenerative Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei, 11031, Taiwan
| | - Wen-Chang Chang
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Yi-Chao Lee
- Ph.D. Program for Neural Regenerative Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei, 11031, Taiwan. .,Center for Neurotrauma and Neuroregeneration, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
21
|
Rahman HNA, Wu H, Dong Y, Pasula S, Wen A, Sun Y, Brophy ML, Tessneer KL, Cai X, McManus J, Chang B, Kwak S, Rahman NS, Xu W, Fernandes C, Mcdaniel JM, Xia L, Smith L, Srinivasan RS, Chen H. Selective Targeting of a Novel Epsin-VEGFR2 Interaction Promotes VEGF-Mediated Angiogenesis. Circ Res 2016; 118:957-969. [PMID: 26879230 DOI: 10.1161/circresaha.115.307679] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Accepted: 02/12/2016] [Indexed: 12/17/2022]
Abstract
RATIONALE We previously reported that vascular endothelial growth factor (VEGF)-induced binding of VEGF receptor 2 (VEGFR2) to epsins 1 and 2 triggers VEGFR2 degradation and attenuates VEGF signaling. The epsin ubiquitin interacting motif (UIM) was shown to be required for the interaction with VEGFR2. However, the molecular determinants that govern how epsin specifically interacts with and regulates VEGFR2 were unknown. OBJECTIVE The goals for the present study were as follows: (1) to identify critical molecular determinants that drive the specificity of the epsin and VEGFR2 interaction and (2) to ascertain whether such determinants were critical for physiological angiogenesis in vivo. METHODS AND RESULTS Structural modeling uncovered 2 novel binding surfaces within VEGFR2 that mediate specific interactions with epsin UIM. Three glutamic acid residues in epsin UIM were found to interact with residues in VEGFR2. Furthermore, we found that the VEGF-induced VEGFR2-epsin interaction promoted casitas B-lineage lymphoma-mediated ubiquitination of epsin, and uncovered a previously unappreciated ubiquitin-binding surface within VEGFR2. Mutational analysis revealed that the VEGFR2-epsin interaction is supported by VEGFR2 interacting specifically with the UIM and with ubiquitinated epsin. An epsin UIM peptide, but not a mutant UIM peptide, potentiated endothelial cell proliferation, migration and angiogenic properties in vitro, increased postnatal retinal angiogenesis, and enhanced VEGF-induced physiological angiogenesis and wound healing. CONCLUSIONS Distinct residues in the epsin UIM and VEGFR2 mediate specific interactions between epsin and VEGFR2, in addition to UIM recognition of ubiquitin moieties on VEGFR2. These novel interactions are critical for pathophysiological angiogenesis, suggesting that these sites could be selectively targeted by therapeutics to modulate angiogenesis.
Collapse
Affiliation(s)
- H N Ashiqur Rahman
- Vascular Biology Program, Karp Family Research Labs #12.214, Harvard Medical School, Boston Children's Hospital, 300 Longwood Ave, Boston, MA 02115, USA
| | - Hao Wu
- Vascular Biology Program, Karp Family Research Labs #12.214, Harvard Medical School, Boston Children's Hospital, 300 Longwood Ave, Boston, MA 02115, USA
| | - Yunzhou Dong
- Vascular Biology Program, Karp Family Research Labs #12.214, Harvard Medical School, Boston Children's Hospital, 300 Longwood Ave, Boston, MA 02115, USA
| | - Satish Pasula
- Cardiovascular Biology Program, Oklahoma Medical Research Foundation, Oklahoma, OK 73104, USA
| | - Aiyun Wen
- Vascular Biology Program, Karp Family Research Labs #12.214, Harvard Medical School, Boston Children's Hospital, 300 Longwood Ave, Boston, MA 02115, USA
| | - Ye Sun
- Vascular Biology Program, Karp Family Research Labs #12.214, Harvard Medical School, Boston Children's Hospital, 300 Longwood Ave, Boston, MA 02115, USA
| | - Megan L Brophy
- Vascular Biology Program, Karp Family Research Labs #12.214, Harvard Medical School, Boston Children's Hospital, 300 Longwood Ave, Boston, MA 02115, USA.,Department of Biochemistry and Molecular Biology, University of Oklahoma Health Science Center, Oklahoma, OK 73104, USA
| | - Kandice L Tessneer
- Cardiovascular Biology Program, Oklahoma Medical Research Foundation, Oklahoma, OK 73104, USA
| | - Xiaofeng Cai
- Vascular Biology Program, Karp Family Research Labs #12.214, Harvard Medical School, Boston Children's Hospital, 300 Longwood Ave, Boston, MA 02115, USA
| | - John McManus
- Cardiovascular Biology Program, Oklahoma Medical Research Foundation, Oklahoma, OK 73104, USA
| | - Baojun Chang
- Cardiovascular Biology Program, Oklahoma Medical Research Foundation, Oklahoma, OK 73104, USA
| | - Sukyoung Kwak
- Vascular Biology Program, Karp Family Research Labs #12.214, Harvard Medical School, Boston Children's Hospital, 300 Longwood Ave, Boston, MA 02115, USA
| | - Negar S Rahman
- Cardiovascular Biology Program, Oklahoma Medical Research Foundation, Oklahoma, OK 73104, USA
| | - Wenjia Xu
- Cardiovascular Biology Program, Oklahoma Medical Research Foundation, Oklahoma, OK 73104, USA
| | - Conrad Fernandes
- Cardiovascular Biology Program, Oklahoma Medical Research Foundation, Oklahoma, OK 73104, USA
| | - John Michael Mcdaniel
- Cardiovascular Biology Program, Oklahoma Medical Research Foundation, Oklahoma, OK 73104, USA
| | - Lijun Xia
- Cardiovascular Biology Program, Oklahoma Medical Research Foundation, Oklahoma, OK 73104, USA
| | - Lois Smith
- Vascular Biology Program, Karp Family Research Labs #12.214, Harvard Medical School, Boston Children's Hospital, 300 Longwood Ave, Boston, MA 02115, USA
| | - R Sathish Srinivasan
- Cardiovascular Biology Program, Oklahoma Medical Research Foundation, Oklahoma, OK 73104, USA
| | - Hong Chen
- Vascular Biology Program, Karp Family Research Labs #12.214, Harvard Medical School, Boston Children's Hospital, 300 Longwood Ave, Boston, MA 02115, USA
| |
Collapse
|
22
|
Peters NC, Berg CA. Dynamin-mediated endocytosis is required for tube closure, cell intercalation, and biased apical expansion during epithelial tubulogenesis in the Drosophila ovary. Dev Biol 2015; 409:39-54. [PMID: 26542010 DOI: 10.1016/j.ydbio.2015.10.034] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2015] [Revised: 08/09/2015] [Accepted: 10/31/2015] [Indexed: 11/28/2022]
Abstract
Most metazoans are able to grow beyond a few hundred cells and to support differentiated tissues because they elaborate multicellular, epithelial tubes that are indispensable for nutrient and gas exchange. To identify and characterize the cellular behaviors and molecular mechanisms required for the morphogenesis of epithelial tubes (i.e., tubulogenesis), we have turned to the D. melanogaster ovary. Here, epithelia surrounding the developing egg chambers first pattern, then form and extend a set of simple, paired, epithelial tubes, the dorsal appendage (DA) tubes, and they create these structures in the absence of cell division or cell death. This genetically tractable system lets us assess the relative contributions that coordinated changes in cell shape, adhesion, orientation, and migration make to basic epithelial tubulogenesis. We find that Dynamin, a conserved regulator of endocytosis and the cytoskeleton, serves a key role in DA tubulogenesis. We demonstrate that Dynamin is required for distinct aspects of DA tubulogenesis: DA-tube closure, DA-tube-cell intercalation, and biased apical-luminal cell expansion. We provide evidence that Dynamin promotes these processes by facilitating endocytosis of cell-cell and cell-matrix adhesion complexes, and we find that precise levels and sub-cellular distribution of E-Cadherin and specific Integrin subunits impact DA tubulogenesis. Thus, our studies identify novel morphogenetic roles (i.e., tube closure and biased apical expansion), and expand upon established roles (i.e., cell intercalation and adhesion remodeling), for Dynamin in tubulogenesis.
Collapse
Affiliation(s)
- Nathaniel C Peters
- University of Washington, Molecular and Cellular Biology Program and Department of Genome Sciences, Box 355065, Seattle, WA 98195-5065, United States
| | - Celeste A Berg
- University of Washington, Molecular and Cellular Biology Program and Department of Genome Sciences, Box 355065, Seattle, WA 98195-5065, United States.
| |
Collapse
|
23
|
Fan F, Ji C, Wu Y, Ferguson SM, Tamarina N, Philipson LH, Lou X. Dynamin 2 regulates biphasic insulin secretion and plasma glucose homeostasis. J Clin Invest 2015; 125:4026-41. [PMID: 26413867 DOI: 10.1172/jci80652] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2014] [Accepted: 08/20/2015] [Indexed: 12/18/2022] Open
Abstract
Alterations in insulin granule exocytosis and endocytosis are paramount to pancreatic β cell dysfunction in diabetes mellitus. Here, using temporally controlled gene ablation specifically in β cells in mice, we identified an essential role of dynamin 2 GTPase in preserving normal biphasic insulin secretion and blood glucose homeostasis. Dynamin 2 deletion in β cells caused glucose intolerance and substantial reduction of the second phase of glucose-stimulated insulin secretion (GSIS); however, mutant β cells still maintained abundant insulin granules, with no signs of cell surface expansion. Compared with control β cells, real-time capacitance measurements demonstrated that exocytosis-endocytosis coupling was less efficient but not abolished; clathrin-mediated endocytosis (CME) was severely impaired at the step of membrane fission, which resulted in accumulation of clathrin-coated endocytic intermediates on the plasma membrane. Moreover, dynamin 2 ablation in β cells led to striking reorganization and enhancement of actin filaments, and insulin granule recruitment and mobilization were impaired at the later stage of GSIS. Together, our results demonstrate that dynamin 2 regulates insulin secretory capacity and dynamics in vivo through a mechanism depending on CME and F-actin remodeling. Moreover, this study indicates a potential pathophysiological link between endocytosis and diabetes mellitus.
Collapse
|
24
|
Hou G, Lou X, Sun Y, Xu S, Zi J, Wang Q, Zhou B, Han B, Wu L, Zhao X, Lin L, Liu S. Biomarker Discovery and Verification of Esophageal Squamous Cell Carcinoma Using Integration of SWATH/MRM. J Proteome Res 2015. [PMID: 26224564 DOI: 10.1021/acs.jproteome.5b00438] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Affiliation(s)
- Guixue Hou
- CAS
Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- Proteomics
Division, BGI-Shenzhen, Shenzhen, Guangdong 518083, China
| | - Xiaomin Lou
- CAS
Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China
| | - Yulin Sun
- National Laboratory of Molecular Oncology, Cancer Institute & Hospital, Chinese Academy of Medical Sciences, 17 Panjiayuan, Chaoyangqu, Beijing 100021, China
| | - Shaohang Xu
- Proteomics
Division, BGI-Shenzhen, Shenzhen, Guangdong 518083, China
| | - Jin Zi
- Proteomics
Division, BGI-Shenzhen, Shenzhen, Guangdong 518083, China
| | - Quanhui Wang
- CAS
Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China
- Proteomics
Division, BGI-Shenzhen, Shenzhen, Guangdong 518083, China
| | - Baojin Zhou
- Proteomics
Division, BGI-Shenzhen, Shenzhen, Guangdong 518083, China
| | - Bo Han
- National Laboratory of Molecular Oncology, Cancer Institute & Hospital, Chinese Academy of Medical Sciences, 17 Panjiayuan, Chaoyangqu, Beijing 100021, China
| | - Lin Wu
- CAS
Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China
| | - Xiaohang Zhao
- National Laboratory of Molecular Oncology, Cancer Institute & Hospital, Chinese Academy of Medical Sciences, 17 Panjiayuan, Chaoyangqu, Beijing 100021, China
| | - Liang Lin
- Proteomics
Division, BGI-Shenzhen, Shenzhen, Guangdong 518083, China
| | - Siqi Liu
- CAS
Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China
- Proteomics
Division, BGI-Shenzhen, Shenzhen, Guangdong 518083, China
| |
Collapse
|
25
|
Chichger H, Braza J, Duong H, Stark M, Harrington EO. Neovascularization in the pulmonary endothelium is regulated by the endosome: Rab4-mediated trafficking and p18-dependent signaling. Am J Physiol Lung Cell Mol Physiol 2015; 309:L700-9. [PMID: 26254426 DOI: 10.1152/ajplung.00235.2015] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Accepted: 08/04/2015] [Indexed: 11/22/2022] Open
Abstract
Neovascularization, the formation of new blood vessels, requires multiple processes including vascular leak, migration, and adhesion. Endosomal proteins, such as Rabs, regulate trafficking of key signaling proteins involved in neovascularization. The novel endosome protein, p18, enhances vascular endothelial (VE)-cadherin recycling from early endosome to cell junction to improve pulmonary endothelial barrier function. Since endothelial barrier integrity is vital in neovascularization, we sought to elucidate the role for endosome proteins p18 and Rab4, Rab7, and Rab9 in the process of vessel formation within the pulmonary vasculature. Overexpression of wild-type p18 (p18(wt)), but not the nonendosomal-binding mutant (p18(N39)), significantly increased lung microvascular endothelial cell migration, adhesion, and both in vitro and in vivo tube formation. Chemical inhibition of mTOR or p38 attenuated the proneovascularization role of p18(wt). Similar to the effect of p18(wt), overexpression of prorecycling wild-type (Rab4(WT)) and endosome-anchored (Rab4(Q67L)) Rab4 enhanced neovascularization processes, whereas molecular inhibition of Rab4, by using the nonendosomal-binding mutant (Rab4(S22N)) attenuated VEGF-induced neovascularization. Unlike p18, Rab4-induced neovascularization was independent of mTOR or p38 inhibition but was dependent on p18 expression. This study shows for the first time that neovascularization within the pulmonary vasculature is dependent on the prorecycling endocytic proteins Rab4 and p18.
Collapse
Affiliation(s)
- Havovi Chichger
- Vascular Research Laboratory, Providence Veterans Affairs Medical Center, and Department of Medicine, Alpert Medical School of Brown University, Providence, Rhode Island
| | - Julie Braza
- Vascular Research Laboratory, Providence Veterans Affairs Medical Center, and Department of Medicine, Alpert Medical School of Brown University, Providence, Rhode Island
| | - Huetran Duong
- Vascular Research Laboratory, Providence Veterans Affairs Medical Center, and Department of Medicine, Alpert Medical School of Brown University, Providence, Rhode Island
| | - Myranda Stark
- Vascular Research Laboratory, Providence Veterans Affairs Medical Center, and Department of Medicine, Alpert Medical School of Brown University, Providence, Rhode Island
| | - Elizabeth O Harrington
- Vascular Research Laboratory, Providence Veterans Affairs Medical Center, and Department of Medicine, Alpert Medical School of Brown University, Providence, Rhode Island
| |
Collapse
|
26
|
Jane-wit D, Surovtseva YV, Qin L, Li G, Liu R, Clark P, Manes TD, Wang C, Kashgarian M, Kirkiles-Smith NC, Tellides G, Pober JS. Complement membrane attack complexes activate noncanonical NF-κB by forming an Akt+ NIK+ signalosome on Rab5+ endosomes. Proc Natl Acad Sci U S A 2015; 112:9686-91. [PMID: 26195760 PMCID: PMC4534258 DOI: 10.1073/pnas.1503535112] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Complement membrane attack complexes (MACs) promote inflammatory functions in endothelial cells (ECs) by stabilizing NF-κB-inducing kinase (NIK) and activating noncanonical NF-κB signaling. Here we report a novel endosome-based signaling complex induced by MACs to stabilize NIK. We found that, in contrast to cytokine-mediated activation, NIK stabilization by MACs did not involve cIAP2 or TRAF3. Informed by a genome-wide siRNA screen, instead this response required internalization of MACs in a clathrin-, AP2-, and dynamin-dependent manner into Rab5(+)endosomes, which recruited activated Akt, stabilized NIK, and led to phosphorylation of IκB kinase (IKK)-α. Active Rab5 was required for recruitment of activated Akt to MAC(+) endosomes, but not for MAC internalization or for Akt activation. Consistent with these in vitro observations, MAC internalization occurred in human coronary ECs in vivo and was similarly required for NIK stabilization and EC activation. We conclude that MACs activate noncanonical NF-κB by forming a novel Akt(+)NIK(+) signalosome on Rab5(+) endosomes.
Collapse
Affiliation(s)
- Dan Jane-wit
- Division of Cardiovascular Medicine, Yale University School of Medicine, New Haven, CT 06520
| | - Yulia V Surovtseva
- Yale Center for Molecular Discovery, Yale University, New Haven, CT 06516
| | - Lingfeng Qin
- Department of Surgery, Yale University School of Medicine, New Haven, CT 06520
| | - Guangxin Li
- Department of Surgery, Yale University School of Medicine, New Haven, CT 06520
| | - Rebecca Liu
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520
| | - Pamela Clark
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520
| | - Thomas D Manes
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520
| | - Chen Wang
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520
| | - Michael Kashgarian
- Department of Pathology, Yale University School of Medicine, New Haven, CT 06520
| | | | - George Tellides
- Department of Surgery, Yale University School of Medicine, New Haven, CT 06520
| | - Jordan S Pober
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520; Department of Pathology, Yale University School of Medicine, New Haven, CT 06520
| |
Collapse
|
27
|
Iwamoto DV, Calderwood DA. Regulation of integrin-mediated adhesions. Curr Opin Cell Biol 2015; 36:41-7. [PMID: 26189062 DOI: 10.1016/j.ceb.2015.06.009] [Citation(s) in RCA: 146] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2015] [Revised: 06/01/2015] [Accepted: 06/30/2015] [Indexed: 11/18/2022]
Abstract
Integrins are heterodimeric transmembrane adhesion receptors that couple the actin cytoskeleton to the extracellular environment and bidirectionally relay signals across the cell membrane. These processes are critical for cell attachment, migration, differentiation, and survival, and therefore play essential roles in metazoan development, physiology, and pathology. Integrin-mediated adhesions are regulated by diverse factors, including the conformation-specific affinities of integrin receptors for their extracellular ligands, the clustering of integrins and their intracellular binding partners into discrete adhesive structures, mechanical forces exerted on the adhesion, and the intracellular trafficking of integrins themselves. Recent advances shed light onto how the interaction of specific intracellular proteins with the short cytoplasmic tails of integrins controls each of these activities.
Collapse
Affiliation(s)
- Daniel V Iwamoto
- Department of Pharmacology, Yale University School of Medicine, 333 Cedar St., New Haven, CT 06520, USA
| | - David A Calderwood
- Department of Pharmacology, Yale University School of Medicine, 333 Cedar St., New Haven, CT 06520, USA; Department of Cell Biology, Yale University School of Medicine, 333 Cedar St., New Haven, CT 06520, USA.
| |
Collapse
|
28
|
Aranda JF, Canfrán-Duque A, Goedeke L, Suárez Y, Fernández-Hernando C. The miR-199-dynamin regulatory axis controls receptor-mediated endocytosis. J Cell Sci 2015; 128:3197-209. [PMID: 26163491 DOI: 10.1242/jcs.165233] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2014] [Accepted: 07/02/2015] [Indexed: 12/19/2022] Open
Abstract
Small non-coding RNAs (microRNAs) are important regulators of gene expression that modulate many physiological processes; however, their role in regulating intracellular transport remains largely unknown. Intriguingly, we found that the dynamin (DNM) genes, a GTPase family of proteins responsible for endocytosis in eukaryotic cells, encode the conserved miR-199a and miR-199b family of miRNAs within their intronic sequences. Here, we demonstrate that miR-199a and miR-199b regulate endocytic transport by controlling the expression of important mediators of endocytosis such as clathrin heavy chain (CLTC), Rab5A, low-density lipoprotein receptor (LDLR) and caveolin-1 (Cav-1). Importantly, miR-199a-5p and miR-199b-5p overexpression markedly inhibits CLTC, Rab5A, LDLR and Cav-1 expression, thus preventing receptor-mediated endocytosis in human cell lines (Huh7 and HeLa). Of note, miR-199a-5p inhibition increases target gene expression and receptor-mediated endocytosis. Taken together, our work identifies a new mechanism by which microRNAs regulate intracellular trafficking. In particular, we demonstrate that the DNM, miR-199a-5p and miR-199b-5p genes act as a bifunctional locus that regulates endocytosis, thus adding an unexpected layer of complexity in the regulation of intracellular trafficking.
Collapse
Affiliation(s)
- Juan F Aranda
- Integrative Cell Signaling and Neurobiology of Metabolism Program, Section of Comparative Medicine, Yale University School of Medicine, New Haven, CT 06510, USA Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Alberto Canfrán-Duque
- Integrative Cell Signaling and Neurobiology of Metabolism Program, Section of Comparative Medicine, Yale University School of Medicine, New Haven, CT 06510, USA Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Leigh Goedeke
- Integrative Cell Signaling and Neurobiology of Metabolism Program, Section of Comparative Medicine, Yale University School of Medicine, New Haven, CT 06510, USA Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Yajaira Suárez
- Integrative Cell Signaling and Neurobiology of Metabolism Program, Section of Comparative Medicine, Yale University School of Medicine, New Haven, CT 06510, USA Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Carlos Fernández-Hernando
- Integrative Cell Signaling and Neurobiology of Metabolism Program, Section of Comparative Medicine, Yale University School of Medicine, New Haven, CT 06510, USA Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT 06510, USA
| |
Collapse
|
29
|
Clegg LW, Mac Gabhann F. Site-Specific Phosphorylation of VEGFR2 Is Mediated by Receptor Trafficking: Insights from a Computational Model. PLoS Comput Biol 2015; 11:e1004158. [PMID: 26067165 PMCID: PMC4466579 DOI: 10.1371/journal.pcbi.1004158] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2014] [Accepted: 01/25/2015] [Indexed: 02/05/2023] Open
Abstract
Matrix-binding isoforms and non-matrix-binding isoforms of vascular endothelial growth factor (VEGF) are both capable of stimulating vascular remodeling, but the resulting blood vessel networks are structurally and functionally different. Here, we develop and validate a computational model of the binding of soluble and immobilized ligands to VEGF receptor 2 (VEGFR2), the endosomal trafficking of VEGFR2, and site-specific VEGFR2 tyrosine phosphorylation to study differences in induced signaling between these VEGF isoforms. In capturing essential features of VEGFR2 signaling and trafficking, our model suggests that VEGFR2 trafficking parameters are largely consistent across multiple endothelial cell lines. Simulations demonstrate distinct localization of VEGFR2 phosphorylated on Y1175 and Y1214. This is the first model to clearly show that differences in site-specific VEGFR2 activation when stimulated with immobilized VEGF compared to soluble VEGF can be accounted for by altered trafficking of VEGFR2 without an intrinsic difference in receptor activation. The model predicts that Neuropilin-1 can induce differences in the surface-to-internal distribution of VEGFR2. Simulations also show that ligated VEGFR2 and phosphorylated VEGFR2 levels diverge over time following stimulation. Using this model, we identify multiple key levers that alter how VEGF binding to VEGFR2 results in different coordinated patterns of multiple downstream signaling pathways. Specifically, simulations predict that VEGF immobilization, interactions with Neuropilin-1, perturbations of VEGFR2 trafficking, and changes in expression or activity of phosphatases acting on VEGFR2 all affect the magnitude, duration, and relative strength of VEGFR2 phosphorylation on tyrosines 1175 and 1214, and they do so predictably within our single consistent model framework. Vascular endothelial growth factor (VEGF) is an important regulator of blood vessel growth. To date, therapies attempting to harness the VEGF system to promote blood vessel growth (e.g. for wound healing or ischemic disease) have achieved only limited success. To improve VEGF-based therapies, we need to better understand how VEGF promotes development of functional blood vessels. We have developed a computational model of VEGF binding to the receptor VEGFR2, trafficking of VEGFR2 through endosomal compartments in the cell, and activation of VEGFR2 on several tyrosine residues. The pattern of tyrosines activated on VEGFR2 influences cell behavior, promoting cell survival, proliferation, or migration. The combination of these cues influences the diameter of vessels, degree of branching, and leakiness of the resultant vessel network. Our model shows that changes in VEGFR2 trafficking as a result of VEGF immobilization to the extracellular matrix are sufficient to describe observed changes in the pattern of VEGFR2 activation compared to stimulation with purely soluble VEGF. This model can be used to predict how VEGF immobilization, interactions with co-receptors or proteins that deactivate VEGFR2, and changes to VEGFR2 trafficking can be tuned to promote development of functional blood vessel networks for tissue engineering applications.
Collapse
Affiliation(s)
- Lindsay Wendel Clegg
- Institute for Computational Medicine and Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland, United States of America
- * E-mail:
| | - Feilim Mac Gabhann
- Institute for Computational Medicine and Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland, United States of America
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, Maryland, United States of America
| |
Collapse
|
30
|
Nelson DA, Larsen M. Heterotypic control of basement membrane dynamics during branching morphogenesis. Dev Biol 2015; 401:103-9. [PMID: 25527075 PMCID: PMC4465071 DOI: 10.1016/j.ydbio.2014.12.011] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2014] [Revised: 11/24/2014] [Accepted: 12/09/2014] [Indexed: 02/06/2023]
Abstract
Many mammalian organs undergo branching morphogenesis to create highly arborized structures with maximized surface area for specialized organ function. Cooperative cell-cell and cell-matrix adhesions that sculpt the emerging tissue architecture are guided by dynamic basement membranes. Properties of the basement membrane are reciprocally controlled by the interacting epithelial and mesenchymal cell populations. Here we discuss how basement membrane remodeling is required for branching morphogenesis to regulate cell-matrix and cell-cell adhesions that are required for cell patterning during morphogenesis and how basement membrane impacts morphogenesis by stimulation of cell patterning, force generation, and mechanotransduction. We suggest that in addition to creating mature epithelial architecture, remodeling of the epithelial basement membrane during branching morphogenesis is also essential to promote maturation of the stromal mesenchyme to create mature organ structure. Recapitulation of developmental cell-matrix and cell-cell interactions are of critical importance in tissue engineering and regeneration strategies that seek to restore organ function.
Collapse
Affiliation(s)
- Deirdre A Nelson
- Department of Biological Sciences, University at Albany, State University of New York, 1400 Washington Avenue, 1400 Washington Ave, Albany, NY 12222, USA
| | - Melinda Larsen
- Department of Biological Sciences, University at Albany, State University of New York, 1400 Washington Avenue, 1400 Washington Ave, Albany, NY 12222, USA.
| |
Collapse
|
31
|
Abstract
Dynamins are highly conserved large GTPases (enzymes that hydrolyze guanosine triphosphate) involved in endocytosis and vesicle transport, and mutations in the ubiquitous and housekeeping dynamin 2 (DNM2) have been associated with thrombocytopenia in humans. To determine the role of DNM2 in thrombopoiesis, we generated Dnm2(fl/fl) Pf4-Cre mice specifically lacking DNM2 in the megakaryocyte (MK) lineage. Dnm2(fl/fl) Pf4-Cre mice had severe macrothrombocytopenia with moderately accelerated platelet clearance. Dnm2-null bone marrow MKs had altered demarcation membrane system formation in vivo due to defective endocytic pathway, and fetal liver-derived Dnm2-null MKs formed proplatelets poorly in vitro, showing that DNM2-dependent endocytosis plays a major role in MK membrane formation and thrombopoiesis. Endocytosis of the thrombopoietin receptor Mpl was impaired in Dnm2-null platelets, causing constitutive phosphorylation of the tyrosine kinase JAK2 and elevated circulating thrombopoietin levels. MK-specific DNM2 deletion severely disrupted bone marrow homeostasis, as reflected by marked expansion of hematopoietic stem and progenitor cells, MK hyperplasia, myelofibrosis, and consequent extramedullary hematopoiesis and splenomegaly. Taken together, our data demonstrate that unrestrained MK growth and proliferation results in rapid myelofibrosis and establishes a previously unrecognized role for DNM2-dependent endocytosis in megakaryopoiesis, thrombopoiesis, and bone marrow homeostasis.
Collapse
|
32
|
Schottenfeld-Roames J, Rosa JB, Ghabrial AS. Seamless tube shape is constrained by endocytosis-dependent regulation of active Moesin. Curr Biol 2014; 24:1756-64. [PMID: 25065756 DOI: 10.1016/j.cub.2014.06.029] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2013] [Revised: 04/03/2014] [Accepted: 06/12/2014] [Indexed: 12/11/2022]
Abstract
Most tubes have seams (intercellular or autocellular junctions that seal membranes together into a tube), but "seamless" tubes also exist. In Drosophila, stellate-shaped tracheal terminal cells make seamless tubes, with single branches running through each of dozens of cellular extensions. We find that mutations in braided impair terminal cell branching and cause formation of seamless tube cysts. We show that braided encodes Syntaxin7 and that cysts also form in cells deficient for other genes required either for membrane scission (shibire) or for early endosome formation (Rab5, Vps45, and Rabenosyn-5). These data define a requirement for early endocytosis in shaping seamless tube lumens. Importantly, apical proteins Crumbs and phospho-Moesin accumulate to aberrantly high levels in braided terminal cells. Overexpression of either Crumbs or phosphomimetic Moesin induced lumenal cysts and decreased terminal branching. Conversely, the braided seamless tube cyst phenotype was suppressed by mutations in crumbs or Moesin. Indeed, mutations in Moesin dominantly suppressed seamless tube cyst formation and restored terminal branching. We propose that early endocytosis maintains normal steady-state levels of Crumbs, which recruits apical phosphorylated (active) Moe, which in turn regulates seamless tube shape through modulation of cortical actin filaments.
Collapse
Affiliation(s)
- Jodi Schottenfeld-Roames
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jeffrey B Rosa
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Amin S Ghabrial
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
33
|
Yaqoob U, Jagavelu K, Shergill U, de Assuncao T, Cao S, Shah VH. FGF21 promotes endothelial cell angiogenesis through a dynamin-2 and Rab5 dependent pathway. PLoS One 2014; 9:e98130. [PMID: 24848261 PMCID: PMC4029959 DOI: 10.1371/journal.pone.0098130] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2014] [Accepted: 04/28/2014] [Indexed: 12/26/2022] Open
Abstract
Binding of angiogenic molecules with cognate receptor tyrosine kinases (RTK) is required for angiogenesis however the precise link between RTK binding, endocytosis, and signaling requires further investigation. Here, we use FGFR1 as a model to test the effects of the large GTPase and endocytosis regulatory molecule dynamin-2 on angiogenic signaling in context of distinct FGF ligands. In vitro, overexpression of dominant negative dynamin-2 (DynK44A) attenuates FGFR1 activation of Erk and tubulogenesis by FGF2. Furthermore, we identify FGF21, a non-classical, FGF ligand implicated in diverse human pathologies as an angiogenic molecule acting through FGFR1 and β-Klotho coreceptor. Disruption of FGFR1 activation of ERK by FGF21 is achieved by perturbation of the function of both dynamin-2 and Rab5 GTPase. In vivo, mice harboring endothelial selective overexpression of DynK44A, show impaired angiogenesis in response to FGF21. In conclusion, dynamin dependent endocytosis of FGFR1 is required for in vitro and in vivo angiogenesis in response to FGF2 and the non-classical FGF ligand, FGF21. These studies extend our understanding of the relationships between RTK binding, internalization, endosomal targeting, and angiogenic signaling.
Collapse
Affiliation(s)
- Usman Yaqoob
- Gastroenterology Research Unit, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Kumaravelu Jagavelu
- Gastroenterology Research Unit, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Uday Shergill
- Gastroenterology Research Unit, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Thiago de Assuncao
- Gastroenterology Research Unit, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Sheng Cao
- Gastroenterology Research Unit, Mayo Clinic, Rochester, Minnesota, United States of America
- * E-mail: (SC); (VHS)
| | - Vijay H. Shah
- Gastroenterology Research Unit, Mayo Clinic, Rochester, Minnesota, United States of America
- * E-mail: (SC); (VHS)
| |
Collapse
|
34
|
Lee MY, Skoura A, Park E, Landskroner-Eiger S, Jozsef L, Luciano AK, Murata T, Pasula S, Dong Y, Bouaouina M, Calderwood DA, Ferguson SM, De Camilli P, Sessa WC. Dynamin 2 regulation of integrin endocytosis, but not VEGF signaling, is crucial for developmental angiogenesis. J Cell Sci 2014. [DOI: 10.1242/jcs.153080] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
35
|
Abstract
Integrins are heterodimeric cell surface adhesion receptors essential for multicellular life. They connect cells to the extracellular environment and transduce chemical and mechanical signals to and from the cell. Intracellular proteins that bind the integrin cytoplasmic tail regulate integrin engagement of extracellular ligands as well as integrin localization and trafficking. Cytoplasmic integrin-binding proteins also function downstream of integrins, mediating links to the cytoskeleton and to signaling cascades that impact cell motility, growth, and survival. Here, we review key integrin-interacting proteins and their roles in regulating integrin activity, localization, and signaling.
Collapse
Affiliation(s)
- Elizabeth M Morse
- Department of Cell Biology and ‡Department of Pharmacology, Yale University School of Medicine , 333 Cedar Street, New Haven, Connecticut 06520, United States
| | | | | |
Collapse
|