1
|
Bayati A, McPherson PS. Alpha-synuclein, autophagy-lysosomal pathway, and Lewy bodies: Mutations, propagation, aggregation, and the formation of inclusions. J Biol Chem 2024; 300:107742. [PMID: 39233232 PMCID: PMC11460475 DOI: 10.1016/j.jbc.2024.107742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 08/21/2024] [Accepted: 08/23/2024] [Indexed: 09/06/2024] Open
Abstract
Research into the pathophysiology of Parkinson's disease (PD) is a fast-paced pursuit, with new findings about PD and other synucleinopathies being made each year. The involvement of various lysosomal proteins, such as TFEB, TMEM175, GBA, and LAMP1/2, marks the rising awareness about the importance of lysosomes in PD and other neurodegenerative disorders. This, along with recent developments regarding the involvement of microglia and the immune system in neurodegenerative diseases, has brought about a new era in neurodegeneration: the role of proinflammatory cytokines on the nervous system, and their downstream effects on mitochondria, lysosomal degradation, and autophagy. More effort is needed to understand the interplay between neuroimmunology and disease mechanisms, as many of the mechanisms remain enigmatic. α-synuclein, a key protein in PD and the main component of Lewy bodies, sits at the nexus between lysosomal degradation, autophagy, cellular stress, neuroimmunology, PD pathophysiology, and disease progression. This review revisits some fundamental knowledge about PD while capturing some of the latest trends in PD research, specifically as it relates to α-synuclein.
Collapse
Affiliation(s)
- Armin Bayati
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill, University, Montreal, Quebec, Canada.
| | - Peter S McPherson
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill, University, Montreal, Quebec, Canada.
| |
Collapse
|
2
|
Afuwape OA, Chanaday NL, Kasap M, Monteggia LM, Kavalali ET. Persistence of quantal synaptic vesicle recycling in virtual absence of dynamins. J Physiol 2024:10.1113/JP286711. [PMID: 39141823 PMCID: PMC11825889 DOI: 10.1113/jp286711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 07/23/2024] [Indexed: 08/16/2024] Open
Abstract
Dynamins are GTPases required for pinching vesicles off the plasma membrane once a critical curvature is reached during endocytosis. Here, we probed dynamin function in central synapses by depleting all three dynamin isoforms in postnatal hippocampal neurons down to negligible levels. We found a decrease in the propensity of evoked neurotransmission as well as a reduction in synaptic vesicle numbers. Recycling of synaptic vesicles during spontaneous or low levels of evoked activity were largely impervious to dynamin depletion, while retrieval of synaptic vesicle components at higher levels of activity was partially arrested. These results suggest the existence of balancing dynamin-independent mechanisms for synaptic vesicle recycling at central synapses. Classical dynamin-dependent mechanisms are not essential for retrieval of synaptic vesicle proteins after quantal single synaptic vesicle fusion, but they become more relevant for membrane retrieval during intense, sustained neuronal activity. KEY POINTS: Loss of dynamin 2 does not impair synaptic transmission. Loss of all three dynamin isoforms mostly affects evoked neurotransmission. Excitatory synapse function is more susceptible to dynamin loss. Spontaneous neurotransmission is only mildly affected by loss of dynamins. Single synaptic vesicle endocytosis is largely dynamin independent.
Collapse
Affiliation(s)
- Olusoji A.T. Afuwape
- Department of Neurosurgery, University of Arkansas for Medical Sciences, 4301 W. Markham street, Little Rock, AR 72205, USA
| | - Natali L. Chanaday
- Department of Physiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Merve Kasap
- U.S. Food and Drug Administration, The Center for Drug Evaluation and Research (CDER), 10903 New Hampshire Ave., Silver spring, MD 20993, USA
| | - Lisa M. Monteggia
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37240-7933, USA
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN 37240-7933, USA
| | - Ege T. Kavalali
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37240-7933, USA
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN 37240-7933, USA
| |
Collapse
|
3
|
Romero MD, Carabeo RA. Dynamin-dependent entry of Chlamydia trachomatis is sequentially regulated by the effectors TarP and TmeA. Nat Commun 2024; 15:4926. [PMID: 38858371 PMCID: PMC11164928 DOI: 10.1038/s41467-024-49350-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 05/30/2024] [Indexed: 06/12/2024] Open
Abstract
Chlamydia invasion of epithelial cells is a pathogen-driven process involving two functionally distinct effectors - TarP and TmeA. They collaborate to promote robust actin dynamics at sites of entry. Here, we extend studies on the molecular mechanism of invasion by implicating the host GTPase dynamin 2 (Dyn2) in the completion of pathogen uptake. Importantly, Dyn2 function is modulated by TarP and TmeA at the levels of recruitment and activation through oligomerization, respectively. TarP-dependent recruitment requires phosphatidylinositol 3-kinase and the small GTPase Rac1, while TmeA has a post-recruitment role related to Dyn2 oligomerization. This is based on the rescue of invasion duration and efficiency in the absence of TmeA by the Dyn2 oligomer-stabilizing small molecule activator Ryngo 1-23. Notably, Dyn2 also regulated turnover of TarP- and TmeA-associated actin networks, with disrupted Dyn2 function resulting in aberrant turnover dynamics, thus establishing the interdependent functional relationship between Dyn2 and the effectors TarP and TmeA.
Collapse
Affiliation(s)
- Matthew D Romero
- Department of Pathology, Microbiology, and Immunology, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Rey A Carabeo
- Department of Pathology, Microbiology, and Immunology, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA.
| |
Collapse
|
4
|
Romero MD, Carabeo RA. Dynamin-dependent entry of Chlamydia trachomatis is sequentially regulated by the effectors TarP and TmeA. RESEARCH SQUARE 2023:rs.3.rs-3376558. [PMID: 37841835 PMCID: PMC10571596 DOI: 10.21203/rs.3.rs-3376558/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/17/2023]
Abstract
Chlamydia invasion of epithelial cells is a pathogen-driven process involving two functionally distinct effectors - TarP and TmeA. They collaborate to promote robust actin dynamics at sites of entry. Here, we extend studies on the molecular mechanism of invasion by implicating the host GTPase dynamin 2 (Dyn2) in the completion of pathogen uptake. Importantly, Dyn2 function is modulated by TarP and TmeA at the levels of recruitment and activation through oligomerization, respectively. TarP-dependent recruitment requires phosphatidylinositol 3-kinase and the small GTPase Rac1, while TmeA has a post-recruitment role related to Dyn2 oligomerization. This is based on the rescue of invasion duration and efficiency in the absence of TmeA by the Dyn2 oligomer-stabilizing small molecule activator Ryngo 1-23. Notably, Dyn2 also regulated turnover of TarP- and TmeA-associated actin networks, with disrupted Dyn2 function resulting in aberrant turnover dynamics, thus establishing the interdependent functional relationship between Dyn2 and the effectors TarP and TmeA.
Collapse
Affiliation(s)
- Matthew D. Romero
- Department of Pathology and Microbiology, College of Medicine, University of Nebraska Medical Center, Omaha, NE
| | - Rey A. Carabeo
- Department of Pathology and Microbiology, College of Medicine, University of Nebraska Medical Center, Omaha, NE
| |
Collapse
|
5
|
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
|
6
|
Trochet D, Bitoun M. A review of Dynamin 2 involvement in cancers highlights a promising therapeutic target. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2021; 40:238. [PMID: 34294140 PMCID: PMC8296698 DOI: 10.1186/s13046-021-02045-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 07/15/2021] [Indexed: 12/23/2022]
Abstract
Dynamin 2 (DNM2) is an ubiquitously expressed large GTPase well known for its role in vesicle formation in endocytosis and intracellular membrane trafficking also acting as a regulator of cytoskeletons. During the last two decades, DNM2 involvement, through mutations or overexpression, emerged in an increasing number of cancers and often associated with poor prognosis. A wide panel of DNM2-dependent processes was described in cancer cells which explains DNM2 contribution to cancer pathomechanisms. First, DNM2 dysfunction may promote cell migration, invasion and metastasis. Second, DNM2 acts on intracellular signaling pathways fostering tumor cell proliferation and survival. Relative to these roles, DNM2 was demonstrated as a therapeutic target able to reduce cell proliferation, induce apoptosis, and reduce the invasive phenotype in a wide range of cancer cells in vitro. Moreover, proofs of concept of therapy by modulation of DNM2 expression was also achieved in vivo in several animal models. Consequently, DNM2 appears as a promising molecular target for the development of anti-invasive agents and the already provided proofs of concept in animal models represent an important step of preclinical development.
Collapse
Affiliation(s)
- Delphine Trochet
- Centre de Recherche en Myologie, Sorbonne Université, Inserm, UMRS 974, Institut de Myologie, F-75013, Paris, France
| | - Marc Bitoun
- Centre de Recherche en Myologie, Sorbonne Université, Inserm, UMRS 974, Institut de Myologie, F-75013, Paris, France.
| |
Collapse
|
7
|
Sajed R, Saeednejad Zanjani L, Rahimi M, Mansoori M, Zarnani AH, Madjd Z, Ghods R. Overexpression and translocation of dynamin 2 promotes tumor aggressiveness in breast carcinomas. EXCLI JOURNAL 2020; 19:1423-1435. [PMID: 33250680 PMCID: PMC7689243 DOI: 10.17179/excli2020-2762] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Accepted: 10/10/2020] [Indexed: 12/14/2022]
Abstract
Dynamin 2 is a GTPase protein that has been implicated in cancer progression through its various roles such as endocytosis, morphogenesis, epithelial-mesenchymal transition (EMT), cellular contractions, and focal adhesion maturation. The increased expression levels of this molecule have been demonstrated with the development of several cancers such as prostate, pancreas, and bladder. However, its clinical significance in breast cancer is unclear yet. In the present study, the membranous, cytoplasmic, and nuclear expression levels of dynamin 2 molecule were evaluated for the first time, using immunohistochemistry (IHC) on tissue microarray (TMA) slides in 113 invasive breast cancer tissues. Moreover, afterward, the association between the dynamin 2 expression and clinicopathological features was determined. Our finding showed that, a higher nuclear expression of dynamin 2 is significantly associated with an increase in tumor stage (P = 0.05), histological grade (P = 0.001), and age of the patients (P = 0.03). In addition, analysis of the cytoplasmic expression levels of this molecule revealed that, there was a statistically significant difference between the expression levels of dynamin 2 among the different breast cancer subtypes (P = 0.003). Moreover, a significant association was found between the increased expression of dynamin 2 membranous and vascular invasion (VI) (P = 0.02). We showed that dynamin 2 protein expression has an association with more aggressive tumor behavior and more advanced disease in the patients with breast cancer; therefore, dynamin 2 molecule could be considered as an indicator of disease progression and aggressiveness.
Collapse
Affiliation(s)
- Roya Sajed
- Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medicine Sciences (IUMS), Tehran, Iran.,Oncopathology Research Center, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | | | - Mandana Rahimi
- Hasheminejad Kidney Center, Pathology Department, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Maryam Mansoori
- Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medicine Sciences (IUMS), Tehran, Iran.,Oncopathology Research Center, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Amir-Hassan Zarnani
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences (TUMS), Tehran, Iran.,Reproductive Immunology Research Center, Avicenna Research Institute (ACECR), Tehran, Iran
| | - Zahra Madjd
- Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medicine Sciences (IUMS), Tehran, Iran.,Oncopathology Research Center, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Roya Ghods
- Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medicine Sciences (IUMS), Tehran, Iran.,Oncopathology Research Center, Iran University of Medical Sciences (IUMS), Tehran, Iran
| |
Collapse
|
8
|
Dynamin regulates the dynamics and mechanical strength of the actin cytoskeleton as a multifilament actin-bundling protein. Nat Cell Biol 2020; 22:674-688. [PMID: 32451441 PMCID: PMC7953826 DOI: 10.1038/s41556-020-0519-7] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Accepted: 04/07/2020] [Indexed: 01/28/2023]
Abstract
The dynamin GTPase is known to bundle actin filaments, but the underlying molecular mechanism and physiological relevance remain unclear. Our genetic analyses revealed a function of dynamin in propelling invasive membrane protrusions during myoblast fusion in vivo. Using biochemistry, total internal reflection fluorescence microscopy, electron microscopy and cryo-electron tomography, we show that dynamin bundles actin while forming a helical structure. At its full capacity, each dynamin helix captures 12-16 actin filaments on the outer rim of the helix. GTP hydrolysis by dynamin triggers disassembly of fully assembled dynamin helices, releasing free dynamin dimers/tetramers and facilitating Arp2/3-mediated branched actin polymerization. The assembly/disassembly cycles of dynamin promote continuous actin bundling to generate mechanically stiff actin super-bundles. Super-resolution and immunogold platinum replica electron microscopy revealed dynamin along actin bundles at the fusogenic synapse. These findings implicate dynamin as a unique multifilament actin-bundling protein that regulates the dynamics and mechanical strength of the actin cytoskeletal network.
Collapse
|
9
|
Dynamins 2 and 3 control the migration of human megakaryocytes by regulating CXCR4 surface expression and ITGB1 activity. Blood Adv 2019; 2:3540-3552. [PMID: 30538113 DOI: 10.1182/bloodadvances.2018021923] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Accepted: 11/04/2018] [Indexed: 12/14/2022] Open
Abstract
Megakaryocyte (MK) migration from the bone marrow periosteal niche toward the vascular niche is a prerequisite for proplatelet extension and release into the circulation. The mechanism for this highly coordinated process is poorly understood. Here we show that dynasore (DNSR), a small-molecule inhibitor of dynamins (DNMs), or short hairpin RNA knockdown of DNM2 and DNM3 impairs directional migration in a human MK cell line or MKs derived from cultured CD34+ cells. Because cell migration requires actin cytoskeletal rearrangements, we measured actin polymerization and the activity of cytoskeleton regulator RhoA and found them to be decreased after inhibition of DNM2 and DNM3. Because SDF-1α is important for hematopoiesis, we studied the expression of its receptor CXCR4 in DNSR-treated cells. CXCR4 expression on the cell surface was increased, at least partially because of slower endocytosis and internalization after SDF-1α treatment. Combined inhibition of DNM2 and DNM3 or forced expression of dominant-negative Dnm2-K44A or GTPase-defective DNM3 diminished β1 integrin (ITGB1) activity. DNSR-treated MKs showed an abnormally clustered staining pattern of Rab11, a marker of recycling endosomes. This suggests decreased recruitment of the recycling pathway in DNSR-treated cells. Altogether, we show that the GTPase activity of DNMs, which governs endocytosis and regulates cell receptor trafficking, exerts control on MK migration toward SDF-1α gradients, such as those originating from the vascular niche. DNMs play a critical role in MKs by triggering membrane-cytoskeleton rearrangements downstream of CXCR4 and integrins.
Collapse
|
10
|
Dynamin-Like Protein B of Dictyostelium Contributes to Cytokinesis Cooperatively with Other Dynamins. Cells 2019; 8:cells8080781. [PMID: 31357517 PMCID: PMC6721605 DOI: 10.3390/cells8080781] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Revised: 07/19/2019] [Accepted: 07/25/2019] [Indexed: 01/31/2023] Open
Abstract
Dynamin is a large GTPase responsible for diverse cellular processes, such as endocytosis, division of organelles, and cytokinesis. The social amoebozoan, Dictyostelium discoideum, has five dynamin-like proteins: dymA, dymB, dlpA, dlpB, and dlpC. DymA, dlpA, or dlpB-deficient cells exhibited defects in cytokinesis. DlpA and dlpB were found to colocalize at cleavage furrows from the early phase, and dymA localized at the intercellular bridge connecting the two daughter cells, indicating that these dynamins contribute to cytokinesis at distinct dividing stages. Total internal reflection fluorescence microscopy revealed that dlpA and dlpB colocalized at individual dots at the furrow cortex. However, dlpA and dlpB did not colocalize with clathrin, suggesting that they are not involved in clathrin-mediated endocytosis. The fact that dlpA did not localize at the furrow in dlpB null cells and vice versa, as well as other several lines of evidence, suggests that hetero-oligomerization of dlpA and dlpB is required for them to bind to the furrow. The hetero-oligomers directly or indirectly associate with actin filaments, stabilizing them in the contractile rings. Interestingly, dlpA, but not dlpB, accumulated at the phagocytic cups independently of dlpB. Our results suggest that the hetero-oligomers of dlpA and dlpB contribute to cytokinesis cooperatively with dymA.
Collapse
|
11
|
Ali T, Bednarska J, Vassilopoulos S, Tran M, Diakonov IA, Ziyadeh-Isleem A, Guicheney P, Gorelik J, Korchev YE, Reilly MM, Bitoun M, Shevchuk A. Correlative SICM-FCM reveals changes in morphology and kinetics of endocytic pits induced by disease-associated mutations in dynamin. FASEB J 2019; 33:8504-8518. [PMID: 31017801 PMCID: PMC6593877 DOI: 10.1096/fj.201802635r] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Dynamin 2 (DNM2) is a GTP-binding protein that controls endocytic vesicle scission and defines a whole class of dynamin-dependent endocytosis, including clathrin-mediated endocytosis by caveoli. It has been suggested that mutations in the DNM2 gene, associated with 3 inherited diseases, disrupt endocytosis. However, how exactly mutations affect the nanoscale morphology of endocytic machinery has never been studied. In this paper, we used live correlative scanning ion conductance microscopy (SICM) and fluorescence confocal microscopy (FCM) to study how disease-associated mutations affect the morphology and kinetics of clathrin-coated pits (CCPs) by directly following their dynamics of formation, maturation, and internalization in skin fibroblasts from patients with centronuclear myopathy (CNM) and in Cos-7 cells expressing corresponding dynamin mutants. Using SICM-FCM, which we have developed, we show how p.R465W mutation disrupts pit structure, preventing its maturation and internalization, and significantly increases the lifetime of CCPs. Differently, p.R522H slows down the formation of CCPs without affecting their internalization. We also found that CNM mutations in DNM2 affect the distribution of caveoli and reduce dorsal ruffling in human skin fibroblasts. Collectively, our SICM-FCM findings at single CCP level, backed up by electron microscopy data, argue for the impairment of several forms of endocytosis in DNM2-linked CNM.-Ali, T., Bednarska, J., Vassilopoulos, S., Tran, M., Diakonov, I. A., Ziyadeh-Isleem, A., Guicheney, P., Gorelik, J., Korchev, Y. E., Reilly, M. M., Bitoun, M., Shevchuk, A. Correlative SICM-FCM reveals changes in morphology and kinetics of endocytic pits induced by disease-associated mutations in dynamin.
Collapse
Affiliation(s)
- Tayyibah Ali
- Division of Experimental Medicine, Department of Medicine, Imperial College London, London, United Kingdom
| | - Joanna Bednarska
- Division of Experimental Medicine, Department of Medicine, Imperial College London, London, United Kingdom
| | - Stéphane Vassilopoulos
- Research Center for Myology, Institut de Myologie, UMRS 974, INSERM, Sorbonne Université, Paris, France
| | - Martin Tran
- Division of Experimental Medicine, Department of Medicine, Imperial College London, London, United Kingdom
| | - Ivan A Diakonov
- Department of Cardiac Medicine, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Azza Ziyadeh-Isleem
- UMRS 1166, INSERM, Institute of Cardiometabolism and Nutrition (ICAN), Paris, France.,UMRS 1166, Sorbonne Universités-Pierre and Marie Curie University (UPMC), Paris, France
| | - Pascale Guicheney
- UMRS 1166, INSERM, Institute of Cardiometabolism and Nutrition (ICAN), Paris, France.,UMRS 1166, Sorbonne Universités-Pierre and Marie Curie University (UPMC), Paris, France
| | - Julia Gorelik
- Department of Cardiac Medicine, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Yuri E Korchev
- Division of Experimental Medicine, Department of Medicine, Imperial College London, London, United Kingdom
| | - Mary M Reilly
- MRC Centre for Neuromuscular Diseases, UCL Institute of Neurology, Queen Square London, United Kingdom
| | - Marc Bitoun
- Research Center for Myology, Institut de Myologie, UMRS 974, INSERM, Sorbonne Université, Paris, France
| | - Andrew Shevchuk
- Division of Experimental Medicine, Department of Medicine, Imperial College London, London, United Kingdom
| |
Collapse
|
12
|
Stevens LM, Moffat K, Cooke L, Nomikou K, Mertens PPC, Jackson T, Darpel KE. A low-passage insect-cell isolate of bluetongue virus uses a macropinocytosis-like entry pathway to infect natural target cells derived from the bovine host. J Gen Virol 2019; 100:568-582. [PMID: 30843784 DOI: 10.1099/jgv.0.001240] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Bluetongue virus (BTV) causes an economically important disease in domestic and wildlife ruminants and is transmitted by Culicoides biting midges. In ruminants, BTV has a wide cell tropism that includes endothelial cells of vascular and lymphatic vessels as important cell targets for virus replication, and several cell types of the immune system including monocytes, macrophages and dendritic cells. Thus, cell-entry represents a particular challenge for BTV as it infects many different cell types in widely diverse vertebrate and invertebrate hosts. Improved understanding of BTV cell-entry could lead to novel antiviral approaches that can block virus transmission from cell to cell between its invertebrate and vertebrate hosts. Here, we have investigated BTV cell-entry using endothelial cells derived from the natural bovine host (BFA cells) and purified whole virus particles of a low-passage, insect-cell isolate of a virulent strain of BTV-1. Our results show that the main entry pathway for infection of BFA cells is dependent on actin and dynamin, and shares certain characteristics with macropinocytosis. The ability to use a macropinocytosis-like entry route could explain the diverse cell tropism of BTV and contribute to the efficiency of transmission between vertebrate and invertebrate hosts.
Collapse
Affiliation(s)
- Lisa M Stevens
- 1The Pirbright Institute, Ash Road, Pirbright, GU24 0NF, UK.,2University of Surrey, Guildford, Surrey, GU2 7XH, UK.,‡Present address: Animal and Plant Health Agency, Woodham Lane, New Haw, KT15 3NB, UK
| | - Katy Moffat
- 1The Pirbright Institute, Ash Road, Pirbright, GU24 0NF, UK
| | - Lyndsay Cooke
- 1The Pirbright Institute, Ash Road, Pirbright, GU24 0NF, UK.,2University of Surrey, Guildford, Surrey, GU2 7XH, UK
| | - Kyriaki Nomikou
- 1The Pirbright Institute, Ash Road, Pirbright, GU24 0NF, UK.,§Present address: School of Veterinary Medicine and Science, University of Nottingham, Sutton Bonnington, Leicestershire, LE12 5RD, UK
| | - Peter P C Mertens
- 1The Pirbright Institute, Ash Road, Pirbright, GU24 0NF, UK.,§Present address: School of Veterinary Medicine and Science, University of Nottingham, Sutton Bonnington, Leicestershire, LE12 5RD, UK
| | - Terry Jackson
- 1The Pirbright Institute, Ash Road, Pirbright, GU24 0NF, UK
| | - Karin E Darpel
- 2University of Surrey, Guildford, Surrey, GU2 7XH, UK.,1The Pirbright Institute, Ash Road, Pirbright, GU24 0NF, UK
| |
Collapse
|
13
|
Gong P, Chen S, Zhang L, Hu Y, Gu A, Zhang J, Wang Y. RhoG-ELMO1-RAC1 is involved in phagocytosis suppressed by mono-butyl phthalate in TM4 cells. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2018; 25:35440-35450. [PMID: 30350139 DOI: 10.1007/s11356-018-3503-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Accepted: 10/16/2018] [Indexed: 06/08/2023]
Abstract
Di-n-butyl phthalate (DBP) is one of the most dominant phthalate esters and is ubiquitous in the environment. Male reproductive toxicity of DBP and its active metabolite mono-butyl phthalate (MBP) has been demonstrated in in vivo and in vitro studies. The objective of this study was to explore the roles of RhoG-ELMO1-RAC1 in phagocytosis disrupted by MBP in TM4 cells. Mouse Sertoli cell lines (TM4 cells) were maintained and treated by various levels of MBP (1, 10, and 100 μM) for 24 h. Then, cells were harvested for further experiments. Phagocytic capacity of TM4 cells was detected by flow cytometry, immunofluorescence, and oil red O staining. RAC1 activity (GTP-RAC1) was measured by RAC1 pull-down assay. Expression of mRNA and protein related to phagocytosis including ELMO1, RhoG, and RAC1 was analyzed by qRT-PCR and Western blots, respectively. MBP inhibited phagocytosis of TM4 cells and downregulated GTP-RAC1 expression and movement to membrane markedly. Furthermore, ELMO1 protein expression was downregulated in a dose-dependent manner after MBP treatments. Additionally, expression of proteins relating to phagocytosis, including RhoG and GTP-RAC1, was decreased significantly, but expression of total-RAC1 remained unchanged. GTP-RAC1 expression increased dramatically after TM4 cells were transfected with ELMO1 or RhoG plasmid, but restored under co-treatments with MBP and ELMO1/RhoG plasmid. This study suggests that MBP can reduce the phagocytosis of Sertoli cells through RhoG-ELMO1-RAC1 pathway.
Collapse
Affiliation(s)
- Pan Gong
- The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166, People's Republic of China
- The Key Laboratory of Reproductive Medicine, Institute of Toxicology, Nanjing Medical University, Nanjing, 211166, People's Republic of China
| | - Shanshan Chen
- The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166, People's Republic of China
- The Key Laboratory of Reproductive Medicine, Institute of Toxicology, Nanjing Medical University, Nanjing, 211166, People's Republic of China
| | - Lulu Zhang
- The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166, People's Republic of China
- The Key Laboratory of Reproductive Medicine, Institute of Toxicology, Nanjing Medical University, Nanjing, 211166, People's Republic of China
- Safety Assessment and Research Center for Drug, Pesticide and Veterinary Drug of Jiangsu Province, Nanjing Medical University, Nanjing, 211166, People's Republic of China
| | - Yanhui Hu
- The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166, People's Republic of China
- The Key Laboratory of Reproductive Medicine, Institute of Toxicology, Nanjing Medical University, Nanjing, 211166, People's Republic of China
- Safety Assessment and Research Center for Drug, Pesticide and Veterinary Drug of Jiangsu Province, Nanjing Medical University, Nanjing, 211166, People's Republic of China
| | - Aihua Gu
- The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166, People's Republic of China
- The Key Laboratory of Reproductive Medicine, Institute of Toxicology, Nanjing Medical University, Nanjing, 211166, People's Republic of China
| | - Jingshu Zhang
- The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166, People's Republic of China
- The Key Laboratory of Reproductive Medicine, Institute of Toxicology, Nanjing Medical University, Nanjing, 211166, People's Republic of China
- Safety Assessment and Research Center for Drug, Pesticide and Veterinary Drug of Jiangsu Province, Nanjing Medical University, Nanjing, 211166, People's Republic of China
| | - Yubang Wang
- The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166, People's Republic of China.
- The Key Laboratory of Reproductive Medicine, Institute of Toxicology, Nanjing Medical University, Nanjing, 211166, People's Republic of China.
- Safety Assessment and Research Center for Drug, Pesticide and Veterinary Drug of Jiangsu Province, Nanjing Medical University, Nanjing, 211166, People's Republic of China.
| |
Collapse
|
14
|
A RAB35-p85/PI3K axis controls oscillatory apical protrusions required for efficient chemotactic migration. Nat Commun 2018; 9:1475. [PMID: 29662076 PMCID: PMC5902610 DOI: 10.1038/s41467-018-03571-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Accepted: 02/15/2018] [Indexed: 11/17/2022] Open
Abstract
How cells move chemotactically remains a major unmet challenge in cell biology. Emerging evidence indicates that for interpreting noisy, shallow gradients of soluble cues a system must behave as an excitable process. Here, through an RNAi-based, high-content screening approach, we identify RAB35 as necessary for the formation of growth factors (GFs)-induced waves of circular dorsal ruffles (CDRs), apically restricted actin-rich migratory protrusions. RAB35 is sufficient to induce recurrent and polarized CDRs that travel as propagating waves, thus behaving as an excitable system that can be biased to control cell steering. Consistently, RAB35 is essential for promoting directed chemotactic migration and chemoinvasion of various cells in response to gradients of motogenic GFs. Molecularly, RAB35 does so by directly regulating the activity of p85/PI3K polarity axis. We propose that RAB35 is a molecular determinant for the control of an excitable, oscillatory system that acts as a steering wheel for GF-mediated chemotaxis and chemoinvasion. Circular dorsal ruffles (CDRs) are apical actin enriched structures involved in the interpretation of growth factor gradients during cell migration. Here, the authors find that a RAB35/PI3K axis is necessary and sufficient for the formation and stabilization of polarized CDRs and persistent directional migration.
Collapse
|
15
|
Yoshida S, Pacitto R, Inoki K, Swanson J. Macropinocytosis, mTORC1 and cellular growth control. Cell Mol Life Sci 2018; 75:1227-1239. [PMID: 29119228 PMCID: PMC5843684 DOI: 10.1007/s00018-017-2710-y] [Citation(s) in RCA: 87] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Revised: 10/31/2017] [Accepted: 11/03/2017] [Indexed: 12/15/2022]
Abstract
The growth and proliferation of metazoan cells are driven by cellular nutrient status and by extracellular growth factors. Growth factor receptors on cell surfaces initiate biochemical signals that increase anabolic metabolism and macropinocytosis, an actin-dependent endocytic process in which relatively large volumes of extracellular solutes and nutrients are internalized and delivered efficiently into lysosomes. Macropinocytosis is prominent in many kinds of cancer cells, and supports the growth of cells transformed by oncogenic K-Ras. Growth factor receptor signaling and the overall metabolic status of the cell are coordinated in the cytoplasm by the mechanistic target-of-rapamycin complex-1 (mTORC1), which positively regulates protein synthesis and negatively regulates molecular salvage pathways such as autophagy. mTORC1 is activated by two distinct Ras-related small GTPases, Rag and Rheb, which associate with lysosomal membranes inside the cell. Rag recruits mTORC1 to the lysosomal surface where Rheb directly binds to and activates mTORC1. Rag is activated by both lysosomal luminal and cytosolic amino acids; Rheb activation requires phosphoinositide 3-kinase, Akt, and the tuberous sclerosis complex-1/2. Signals for activation of Rag and Rheb converge at the lysosomal membrane, and several lines of evidence support the idea that growth factor-dependent endocytosis facilitates amino acid transfer into the lysosome leading to the activation of Rag. This review summarizes evidence that growth factor-stimulated macropinocytosis is essential for amino acid-dependent activation of mTORC1, and that increased solute accumulation by macropinocytosis in transformed cells supports unchecked cell growth.
Collapse
Affiliation(s)
- Sei Yoshida
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI, 48109-5620, USA
| | - Regina Pacitto
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI, 48109-5620, USA
| | - Ken Inoki
- Department of Integrative and Molecular Physiology and Internal Medicine, Life Sciences Institute, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Joel Swanson
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI, 48109-5620, USA.
| |
Collapse
|
16
|
Caldas LA, Soares LL, Henrique Seabra S, Attias M, de Souza W. Monitoring of dynamin during the Toxoplasma gondii cell cycle. Pathog Dis 2016; 74:ftw108. [PMID: 27811048 DOI: 10.1093/femspd/ftw108] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Revised: 05/29/2016] [Accepted: 10/29/2016] [Indexed: 11/13/2022] Open
Abstract
The obligate intracellular protozoan parasite Toxoplasma gondii actively invades virtually all warm-blooded nucleated cells. This process results in a non-fusogenic vacuole, inside which the parasites replicate continuously until egress signaling is triggered. In this work, we investigated the role of the large GTPase dynamin in the interaction of T. gondii with the host cell by using laser and electron microscopy during three key stages: invasion, development and egress. The detection of dynamin during invasion indicates the occurrence of endocytosis, while T. gondii egress appeared to be independent of dynamin participation. However, the presence of dynamin during T. gondii development suggests that this molecule plays undescribed roles in the tachyzoite's cell cycle.
Collapse
Affiliation(s)
- Lucio Ayres Caldas
- Instituto de Biofí-sica, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ 21941-902, Brazil
| | - Leandro Lemgruber Soares
- Instituto Nacional de Metrologia Normalizacao e Qualidade Industrial, Duque de Caxias, RJ 25250-020, Brazil.,Wellcome Trust Centre for Molecular Parasitology, University of Glasgow, 120 University Place Glasgow, Glasgow G12 8QQ, UK
| | - Sergio Henrique Seabra
- Microbiologia, UEZO, Av. Manuel Caldeira de Alvarenga, Rio de Janeiro, RJ 23070-200, Brazil
| | - Marcia Attias
- Instituto de Biofí-sica, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ 21941-902, Brazil
| | - Wanderley de Souza
- Instituto de Biofí-sica, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ 21941-902, Brazil
| |
Collapse
|
17
|
Pal K, Hwang SH, Somatilaka B, Badgandi H, Jackson PK, DeFea K, Mukhopadhyay S. Smoothened determines β-arrestin-mediated removal of the G protein-coupled receptor Gpr161 from the primary cilium. J Cell Biol 2016; 212:861-75. [PMID: 27002170 PMCID: PMC4810300 DOI: 10.1083/jcb.201506132] [Citation(s) in RCA: 112] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Accepted: 02/11/2016] [Indexed: 02/08/2023] Open
Abstract
Dynamic changes in membrane protein composition of the primary cilium are central to development and homeostasis, but we know little about mechanisms regulating membrane protein flux. Stimulation of the sonic hedgehog (Shh) pathway in vertebrates results in accumulation and activation of the effector Smoothened within cilia and concomitant disappearance of a negative regulator, the orphan G protein-coupled receptor (GPCR), Gpr161. Here, we describe a two-step process determining removal of Gpr161 from cilia. The first step involves β-arrestin recruitment by the signaling competent receptor, which is facilitated by the GPCR kinase Grk2. An essential factor here is the ciliary trafficking and activation of Smoothened, which by increasing Gpr161-β-arrestin binding promotes Gpr161 removal, both during resting conditions and upon Shh pathway activation. The second step involves clathrin-mediated endocytosis, which functions outside of the ciliary compartment in coordinating Gpr161 removal. Mechanisms determining dynamic compartmentalization of Gpr161 in cilia define a new paradigm for down-regulation of GPCRs during developmental signaling from a specialized subcellular compartment.
Collapse
Affiliation(s)
- Kasturi Pal
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Sun-Hee Hwang
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Bandarigoda Somatilaka
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Hemant Badgandi
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Peter K Jackson
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305
| | - Kathryn DeFea
- Division of Biomedical Sciences, University of California, Riverside, Riverside, CA 92521
| | - Saikat Mukhopadhyay
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390
| |
Collapse
|
18
|
Hetzenecker S, Helenius A, Krzyzaniak MA. HCMV Induces Macropinocytosis for Host Cell Entry in Fibroblasts. Traffic 2016; 17:351-68. [PMID: 26650385 DOI: 10.1111/tra.12355] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2015] [Revised: 11/30/2015] [Accepted: 11/30/2015] [Indexed: 12/14/2022]
Abstract
Human cytomegalovirus (HCMV) is an important and widespread pathogen in the human population. While infection by this β-herpesvirus in endothelial, epithelial and dendritic cells depends on endocytosis, its entry into fibroblasts is thought to occur by direct fusion of the viral envelope with the plasma membrane. To characterize individual steps during entry in primary human fibroblasts, we employed quantitative assays as well as electron, fluorescence and live cell microscopy in combination with a variety of inhibitory compounds. Our results showed that while infectious entry was pH- and clathrin-independent, it required multiple, endocytosis-related factors and processes. The virions were found to undergo rapid internalization into large vacuoles containing internalized fluid and endosome markers. The characteristics of the internalization process fulfilled major criteria for macropinocytosis. Moreover, we found that soon after addition to fibroblasts the virus rapidly triggered the formation of circular dorsal ruffles in the host cell followed by the generation of large macropinocytic vacuoles. This distinctive form of macropinocytosis has been observed especially in primary cells but has not previously been reported in response to virus stimulation.
Collapse
Affiliation(s)
| | - Ari Helenius
- Institute of Biochemistry, ETH Zurich, Zurich 8093, Switzerland
| | - Magdalena Anna Krzyzaniak
- Institute of Biochemistry, ETH Zurich, Zurich 8093, Switzerland.,Department of Biomedicine, University of Basel, Basel 40001, Switzerland
| |
Collapse
|
19
|
Han SC, Guo HC, Sun SQ, Jin Y, Wei YQ, Feng X, Yao XP, Cao SZ, Xiang Liu D, Liu XT. Productive Entry of Foot-and-Mouth Disease Virus via Macropinocytosis Independent of Phosphatidylinositol 3-Kinase. Sci Rep 2016; 6:19294. [PMID: 26757826 PMCID: PMC4725844 DOI: 10.1038/srep19294] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Accepted: 12/08/2015] [Indexed: 12/12/2022] Open
Abstract
Virus entry is an attractive target for therapeutic intervention. Here, using a combination of electron microscopy, immunofluorescence assay, siRNA interference, specific pharmacological inhibitors, and dominant negative mutation, we demonstrated that the entry of foot-and-mouth disease virus (FMDV) triggered a substantial amount of plasma membrane ruffling. We also found that the internalization of FMDV induced a robust increase in fluid-phase uptake, and virions internalized within macropinosomes colocalized with phase uptake marker dextran. During this stage, the Rac1-Pak1 signaling pathway was activated. After specific inhibition on actin, Na(+)/H(+) exchanger, receptor tyrosine kinase, Rac1, Pak1, myosin II, and protein kinase C, the entry and infection of FMDV significantly decreased. However, inhibition of phosphatidylinositol 3-kinase (PI3K) did not reduce FMDV internalization but increased the viral entry and infection to a certain extent, implying that FMDV entry did not require PI3K activity. Results showed that internalization of FMDV exhibited the main hallmarks of macropinocytosis. Moreover, intracellular trafficking of FMDV involves EEA1/Rab5-positive vesicles. The present study demonstrated macropinocytosis as another endocytic pathway apart from the clathrin-mediated pathway. The findings greatly expand our understanding of the molecular mechanisms of FMDV entry into cells, as well as provide potential insights into the entry mechanisms of other picornaviruses.
Collapse
Affiliation(s)
- Shi-Chong Han
- State Key Laboratory of Veterinary Etiological Biology and OIE/National Foot and Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
| | - Hui-Chen Guo
- State Key Laboratory of Veterinary Etiological Biology and OIE/National Foot and Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
| | - Shi-Qi Sun
- State Key Laboratory of Veterinary Etiological Biology and OIE/National Foot and Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
| | - Ye Jin
- State Key Laboratory of Veterinary Etiological Biology and OIE/National Foot and Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
| | - Yan-Quan Wei
- State Key Laboratory of Veterinary Etiological Biology and OIE/National Foot and Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
| | - Xia Feng
- State Key Laboratory of Veterinary Etiological Biology and OIE/National Foot and Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
| | - Xue-Ping Yao
- College of Veterinary Medicine, Sichuan Agricultural University, Ya’an, Sichuan, China
| | - Sui-Zhong Cao
- College of Veterinary Medicine, Sichuan Agricultural University, Ya’an, Sichuan, China
| | - Ding Xiang Liu
- State Key Laboratory of Veterinary Etiological Biology and OIE/National Foot and Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
- School of Biological Sciences, Nanyang Technological University, Singapore
| | - Xiang-Tao Liu
- State Key Laboratory of Veterinary Etiological Biology and OIE/National Foot and Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
| |
Collapse
|
20
|
Alexandrova AY. Plasticity of tumor cell migration: acquisition of new properties or return to the past? BIOCHEMISTRY (MOSCOW) 2015; 79:947-63. [PMID: 25385021 DOI: 10.1134/s0006297914090107] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
During tumor development cancer cells pass through several stages when cell morphology and migration abilities change remarkably. These stages are named epithelial-mesenchymal and mesenchymal-amoeboid transitions. The molecular mechanisms underlying cell motility are changing during these transitions. As result of transitions the cells acquire new characteristics and modes of motility. Cell migration becomes more independent from the environmental conditions, and thus cell dissemination becomes more aggressive, which leads to formation of distant metastases. In this review we discuss the characteristics of each of the transitions, cell morphology, and the specificity of cellular structures responsible for different modes of cell motility as well as molecular mechanisms regulating each transition.
Collapse
Affiliation(s)
- A Y Alexandrova
- Institute of Carcinogenesis, Blokhin Cancer Research Center, Russian Academy of Medical Sciences, Moscow, 115478, Russia.
| |
Collapse
|
21
|
Lee CS, Kim JM, Ghim J, Suh PG, Ryu SH. GTP-dependent interaction between phospholipase D and dynamin modulates fibronectin-induced cell spreading. Cell Signal 2015; 27:2363-70. [PMID: 26341143 DOI: 10.1016/j.cellsig.2015.08.019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Accepted: 08/30/2015] [Indexed: 11/27/2022]
Abstract
Phospholipase D (PLD) is one of the key enzymes to mediate a variety of cellular phenomena including endocytosis, actin rearrangement, proliferation, differentiation, and migration. Dynamin as a PLD-interacting partner is a large GTP binding protein that has been considered a mechanochemical enzyme involved in endocytosis by hydrolyzing GTP. Although both PLD and dynamin have been implicated in the regulation of actin cytoskeleton, it is not known how they have a link to regulate fibronectin (FN)-induced cell spreading. Furthermore, it is unknown whether dynamin can work as a GTP-dependent regulator through its interaction with other proteins. Here, we demonstrate that PLD can be regulated by dynamin in a GTP-dependent manner and that this is critical for FN-mediated cell spreading. First, we verified that GTP-loaded dynamin can mediate the cell spreading by FN by using dynamin's GTP binding deficient mutant (K44A). Also, we confirmed that blocking the PLD activity inhibited FN-induced cell spreading, not cell adhesion. Moreover, PLD interacted with dynamin in a GTP-dependent manner in FN signaling, and this interaction was crucial for FN-induced PLD activation and cell spreading. Also, we found that PLD mutant (R128K) that didn't have GAP activity increased the GTP-dependent interaction between PLD and dynamin; it also increased PLD activity and cell spreading. These findings suggest that the observed increase in PLD activity was through boosting the binding of PLD with dynamin and it facilitated FN-induced cell spreading. These results imply that GTP-loaded dynamin, like a small GTPase could mediate a "switch on" signaling via interaction with PLD that has a role as an effector.
Collapse
Affiliation(s)
- Chang Sup Lee
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang 790-784, South Korea
| | - Jong Min Kim
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang 790-784, South Korea
| | - Jaewang Ghim
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang 790-784, South Korea
| | - Pann-Ghill Suh
- School of Life Science, Ulsan National Institute of Science and Technology (UNIST), Ulsan 689-798, South Korea
| | - Sung Ho Ryu
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang 790-784, South Korea.
| |
Collapse
|
22
|
Li N, Li S. Epigenetic inactivation of SOX1 promotes cell migration in lung cancer. Tumour Biol 2015; 36:4603-10. [DOI: 10.1007/s13277-015-3107-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2014] [Accepted: 01/14/2015] [Indexed: 11/28/2022] Open
|
23
|
Abstract
UNLABELLED The entry mechanism of murine amphotropic retrovirus (A-MLV) has not been unambiguously determined. We show here that A-MLV is internalized not by caveolae or other pinocytic mechanisms but by macropinocytosis. Thus, A-MLV infection of mouse embryonic fibroblasts deficient for caveolin or dynamin, and NIH 3T3 cells knocked down for caveolin expression, was unaffected. Conversely, A-MLV infection of NIH 3T3 and HeLa cells was sensitive to amiloride analogues and actin-depolymerizing drugs that interfere with macropinocytosis. Further manipulation of the actin cytoskeleton through conditional expression of dominant positive or negative mutants of Rac1, PAK1, and RhoG, to increase or decrease macropinocytosis, similarly correlated with an augmented or inhibited infection with A-MLV, respectively. The same experimental perturbations affected the infection of viruses that use clathrin-coated-pit endocytosis or other pathways for entry only mildly or not at all. These data agree with immunofluorescence studies and cryo-immunogold labeling for electron microscopy, which demonstrate the presence of A-MLV in protrusion-rich areas of the cell surface and in cortical fluid phase (dextran)-filled macropinosomes, which also account for up to a half of the cellular uptake of the cell surface-binding lectin concanavalin A. We conclude that A-MLV use macropinocytosis as the predominant entry portal into cells. IMPORTANCE Binding and entry of virus particles into mammalian cells are the first steps of infection. Understanding how pathogens and toxins exploit or divert endocytosis pathways has advanced our understanding of membrane trafficking pathways, which benefits development of new therapeutic schemes and methods of drug delivery. We show here that amphotropic murine leukemia virus (A-MLV) pseudotyped with the amphotropic envelope protein (which expands the host range to many mammalian cells) gains entry into host cells by macropinocytosis. Macropinosomes form as large, fluid-filled vacuoles (up to 10 μm) following the collapse of cell surface protrusions and membrane scission. We used drugs or the introduction of mutant proteins that affect the actin cytoskeleton and cell surface dynamics to show that macropinocytosis and A-MLV infection are correlated, and we provide both light- and electron-microscopic evidence to show the localization of A-MLV in macropinosomes. Finally, we specifically exclude some other potential entry portals, including caveolae, previously suggested to internalize A-MLV.
Collapse
|
24
|
Hofmann D, Tenzer S, Bannwarth MB, Messerschmidt C, Glaser SF, Schild H, Landfester K, Mailänder V. Mass spectrometry and imaging analysis of nanoparticle-containing vesicles provide a mechanistic insight into cellular trafficking. ACS NANO 2014; 8:10077-10088. [PMID: 25244389 DOI: 10.1021/nn502754c] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Rational design of nanocarriers for drug delivery approaches requires an unbiased knowledge of uptake mechanisms and intracellular trafficking pathways. Here we dissected these processes using a quantitative proteomics approach. We isolated intracellular vesicles containing superparamagnetic iron oxide polystyrene nanoparticles and analyzed their protein composition by label-free quantitative mass spectrometry. The proteomic snapshot of organelle marker proteins revealed that an atypical macropinocytic-like mechanism mediated the entry of nanoparticles. We show that the entry mechanism is controlled by actin reorganization, atypical macropinocytic signaling, and ADP-ribosylation factor 1. Additionally, our proteomics data demonstrated a central role for multivesicular bodies and multilamellar lysosomes in trafficking and final nanoparticle storage. This was confirmed by confocal microscopy and cryo-TEM measurements. By quantitatively analyzing the protein composition of nanoparticle-containing vesicles, our study clearly defines the routes of nanoparticle entry, intracellular trafficking, and the proteomic milieu of a nanoparticle-containing vesicle.
Collapse
Affiliation(s)
- Daniel Hofmann
- Max Planck Institute for Polymer Research , Ackermannweg 10, 55128 Mainz, Germany
| | | | | | | | | | | | | | | |
Collapse
|
25
|
Shin NY, Choi H, Neff L, Wu Y, Saito H, Ferguson SM, De Camilli P, Baron R. Dynamin and endocytosis are required for the fusion of osteoclasts and myoblasts. ACTA ACUST UNITED AC 2014; 207:73-89. [PMID: 25287300 PMCID: PMC4195819 DOI: 10.1083/jcb.201401137] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Dynamin function is essential for cell–cell fusion in both osteoclast precursors and myoblasts in part through its effects on endocytosis. Cell–cell fusion is an evolutionarily conserved process that leads to the formation of multinucleated myofibers, syncytiotrophoblasts and osteoclasts, allowing their respective functions. Although cell–cell fusion requires the presence of fusogenic membrane proteins and actin-dependent cytoskeletal reorganization, the precise machinery allowing cells to fuse is still poorly understood. Using an inducible knockout mouse model to generate dynamin 1– and 2–deficient primary osteoclast precursors and myoblasts, we found that fusion of both cell types requires dynamin. Osteoclast and myoblast cell–cell fusion involves the formation of actin-rich protrusions closely associated with clathrin-mediated endocytosis in the apposed cell. Furthermore, impairing endocytosis independently of dynamin also prevented cell–cell fusion. Since dynamin is involved in both the formation of actin-rich structures and in endocytosis, our results indicate that dynamin function is central to the osteoclast precursors and myoblasts fusion process, and point to an important role of endocytosis in cell–cell fusion.
Collapse
Affiliation(s)
- Nah-Young Shin
- Department of Medicine, Harvard Medical School, Boston, MA 02115 Department of Oral Medicine, Infection and Immunity, Harvard School of Dental Medicine, Boston, MA 02115
| | - Hyewon Choi
- Department of Medicine, Harvard Medical School, Boston, MA 02115 Department of Oral Medicine, Infection and Immunity, Harvard School of Dental Medicine, Boston, MA 02115
| | - Lynn Neff
- Department of Medicine, Harvard Medical School, Boston, MA 02115 Department of Oral Medicine, Infection and Immunity, Harvard School of Dental Medicine, Boston, MA 02115
| | - Yumei Wu
- Department of Cell Biology and Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale School of Medicine, New Haven, CT 06510 Department of Cell Biology and Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale School of Medicine, New Haven, CT 06510
| | - Hiroaki Saito
- Department of Medicine, Harvard Medical School, Boston, MA 02115 Department of Oral Medicine, Infection and Immunity, Harvard School of Dental Medicine, Boston, MA 02115
| | - Shawn M Ferguson
- Department of Cell Biology and Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale School of Medicine, New Haven, CT 06510 Department of Cell Biology and Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale School of Medicine, New Haven, CT 06510
| | - Pietro De Camilli
- Department of Cell Biology and Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale School of Medicine, New Haven, CT 06510 Department of Cell Biology and Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale School of Medicine, New Haven, CT 06510 Howard Hughes Medical Institute, Chevy Chase, MD 20815
| | - Roland Baron
- Department of Medicine, Harvard Medical School, Boston, MA 02115 Department of Oral Medicine, Infection and Immunity, Harvard School of Dental Medicine, Boston, MA 02115
| |
Collapse
|
26
|
Chou AM, Sem KP, Wright GD, Sudhaharan T, Ahmed S. Dynamin1 is a novel target for IRSp53 protein and works with mammalian enabled (Mena) protein and Eps8 to regulate filopodial dynamics. J Biol Chem 2014; 289:24383-96. [PMID: 25031323 DOI: 10.1074/jbc.m114.553883] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Filopodia are dynamic actin-based structures that play roles in processes such as cell migration, wound healing, and axonal guidance. Cdc42 induces filopodial formation through IRSp53, an Inverse-Bin-Amphiphysins-Rvs (I-BAR) domain protein. Previous work from a number of laboratories has shown that IRSp53 generates filopodia by coupling membrane protrusion with actin dynamics through its Src homology 3 domain binding partners. Here, we show that dynamin1 (Dyn1), the large guanosine triphosphatase, is an interacting partner of IRSp53 through pulldown and Förster resonance energy transfer analysis, and we explore its role in filopodial formation. In neuroblastoma cells, Dyn1 localizes to filopodia, associated tip complexes, and the leading edge just behind the anti-capping protein mammalian enabled (Mena). Dyn1 knockdown reduces filopodial formation, which can be rescued by overexpressing wild-type Dyn1 but not the GTPase mutant Dyn1-K44A and the loss-of-function actin binding domain mutant Dyn1-K/E. Interestingly, dynasore, an inhibitor of Dyn GTPase, also reduced filopodial number and increased their lifetime. Using rapid time-lapse total internal reflection fluorescence microscopy, we show that Dyn1 and Mena localize to filopodia only during initiation and assembly. Dyn1 actin binding domain mutant inhibits filopodial formation, suggesting a role in actin elongation. In contrast, Eps8, an actin capping protein, is seen most strongly at filopodial tips during disassembly. Taken together, the results suggest IRSp53 partners with Dyn1, Mena, and Eps8 to regulate filopodial dynamics.
Collapse
Affiliation(s)
- Ai Mei Chou
- From the Institute of Medical Biology, Immunos, 8A Biomedical Grove, Singapore 138648, Singapore
| | - Kai Ping Sem
- From the Institute of Medical Biology, Immunos, 8A Biomedical Grove, Singapore 138648, Singapore
| | - Graham Daniel Wright
- From the Institute of Medical Biology, Immunos, 8A Biomedical Grove, Singapore 138648, Singapore
| | - Thankiah Sudhaharan
- From the Institute of Medical Biology, Immunos, 8A Biomedical Grove, Singapore 138648, Singapore
| | - Sohail Ahmed
- From the Institute of Medical Biology, Immunos, 8A Biomedical Grove, Singapore 138648, Singapore
| |
Collapse
|
27
|
Morton PE, Hicks A, Nastos T, Santis G, Parsons M. CAR regulates epithelial cell junction stability through control of E-cadherin trafficking. Sci Rep 2013; 3:2889. [PMID: 24096322 PMCID: PMC3791454 DOI: 10.1038/srep02889] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2013] [Accepted: 09/20/2013] [Indexed: 12/13/2022] Open
Abstract
CAR (Coxsackie and Adenovirus Receptor) is the primary docking receptor for typeB coxsackie viruses and subgroup C adenoviruses. CAR is a member of the JAM family of adhesion receptors and is located to both tight and adherens junctions between epithelial cells where it can assemble adhesive contacts through homodimerisation in trans. However, the role of CAR in controlling epithelial junction dynamics remains poorly understood. Here we demonstrate that levels of CAR in human epithelial cells play a key role in determining epithelial cell adhesion through control of E-cadherin stability at cell-cell junctions. Mechanistically, we show that CAR is phosphorylated within the C-terminus by PKCδ and that this in turn controls Src-dependent endocytosis of E-cadherin at cell junctions. This data demonstrates a novel role for CAR in regulating epithelial homeostasis.
Collapse
Affiliation(s)
- Penny E Morton
- 1] Division of Asthma, Allergy & Lung Biology, King's College London, 5th Floor Tower Wing, Guy's Hospital Campus, London, United Kingdom [2] Randall Division of Cell and Molecular Biophysics, King's College London, New Hunt's House, Guys Campus, London, United Kingdom
| | | | | | | | | |
Collapse
|
28
|
Sever S, Chang J, Gu C. Dynamin rings: not just for fission. Traffic 2013; 14:1194-9. [PMID: 23980695 PMCID: PMC3830594 DOI: 10.1111/tra.12116] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2013] [Revised: 08/22/2013] [Accepted: 08/26/2013] [Indexed: 12/21/2022]
Abstract
The GTPase dynamin has captivated researchers for over two decades, even managing to establish its own research field. Dynamin's allure is partly due to its unusual biochemical properties as well as its essential role in multiple cellular processes, which include the regulation of clathrin-mediated endocytosis and of actin cytoskeleton. On the basis of the classic model, dynamin oligomerization into higher order oligomers such as rings and helices directly executes the final fission reaction in endocytosis, which results in the generation of clathrin-coated vesicles. Dynamin's role in the regulation of actin cytoskeleton is mostly explained by its interactions with a number of actin-binding and -regulating proteins; however, the molecular mechanism of dynamin's action continues to elude us. Recent insights into the mechanism and role of dynamin oligomerization in the regulation of actin polymerization point to a novel role for dynamin oligomerization in the cell.
Collapse
Affiliation(s)
- Sanja Sever
- Nephrology Division, Massachusetts General Hospital, CNY 149 8.113, 149 13th Street, Charlestown, MA, 02129, USA
| | | | | |
Collapse
|
29
|
Park RJ, Shen H, Liu L, Liu X, Ferguson SM, De Camilli P. Dynamin triple knockout cells reveal off target effects of commonly used dynamin inhibitors. J Cell Sci 2013; 126:5305-12. [PMID: 24046449 DOI: 10.1242/jcs.138578] [Citation(s) in RCA: 179] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Dynamin, which is encoded by three genes in mammals, is a GTPase implicated in endocytic membrane fission. Dynamin 1 and 3 are predominantly expressed in brain, whereas dynamin 2 is ubiquitously expressed. With the goal of assessing the impact of the lack of dynamin on cell physiology, we previously generated and characterized dynamin 1 and 2 double knockout (DKO) fibroblasts. These DKO cells were unexpectedly viable in spite of a severe impairment of clathrin-mediated endocytosis. As low-level expression of the dynamin 3 gene in these cells could not be excluded, we have now engineered dynamin 1, 2 and 3 triple KO (TKO) fibroblasts. These cells did not reveal any additional defects beyond what was previously observed in DKO fibroblasts. Surprisingly, although fluid-phase endocytosis and peripheral membrane ruffling were not impaired by the lack of all three dynamins, two structurally similar, widely used dynamin inhibitors, dynasore and Dyngo-4a, robustly inhibited these two processes both in wild-type and TKO cells. Dynamin TKO cells will be useful tools for the further exploration of dynamin-dependent processes and the development of more specific dynamin inhibitors.
Collapse
Affiliation(s)
- Ryan J Park
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT 06510, USA
| | | | | | | | | | | |
Collapse
|
30
|
Tugizov SM, Herrera R, Palefsky JM. Epstein-Barr virus transcytosis through polarized oral epithelial cells. J Virol 2013; 87:8179-8194. [PMID: 23698302 PMCID: PMC3700193 DOI: 10.1128/jvi.00443-13] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2013] [Accepted: 04/29/2013] [Indexed: 01/28/2023] Open
Abstract
Although Epstein-Barr virus (EBV) is an orally transmitted virus, viral transmission through the oropharyngeal mucosal epithelium is not well understood. In this study, we investigated how EBV traverses polarized human oral epithelial cells without causing productive infection. We found that EBV may be transcytosed through oral epithelial cells bidirectionally, from both the apical to the basolateral membranes and the basolateral to the apical membranes. Apical to basolateral EBV transcytosis was substantially reduced by amiloride, an inhibitor of macropinocytosis. Electron microscopy showed that virions were surrounded by apical surface protrusions and that virus was present in subapical vesicles. Inactivation of signaling molecules critical for macropinocytosis, including phosphatidylinositol 3-kinases, myosin light-chain kinase, Ras-related C3 botulinum toxin substrate 1, p21-activated kinase 1, ADP-ribosylation factor 6, and cell division control protein 42 homolog, led to significant reduction in EBV apical to basolateral transcytosis. In contrast, basolateral to apical EBV transcytosis was substantially reduced by nystatin, an inhibitor of caveolin-mediated virus entry. Caveolae were detected in the basolateral membranes of polarized human oral epithelial cells, and virions were detected in caveosome-like endosomes. Methyl β-cyclodextrin, an inhibitor of caveola formation, reduced EBV basolateral entry. EBV virions transcytosed in either direction were able to infect B lymphocytes. Together, these data show that EBV transmigrates across oral epithelial cells by (i) apical to basolateral transcytosis, potentially contributing to initial EBV penetration that leads to systemic infection, and (ii) basolateral to apical transcytosis, which may enable EBV secretion into saliva in EBV-infected individuals.
Collapse
Affiliation(s)
- Sharof M Tugizov
- Department of Medicine, University of California San Francisco, San Francisco, California, USA.
| | | | | |
Collapse
|
31
|
Abstract
Cytoskeletal dynamics are key to the establishment of cell polarity and the consequent coordination of protrusion and contraction that drives cell migration. During these events, the actin and microtubule cytoskeleton act in concert with the cellular machinery that controls endo-and exocytosis, thus regulating polarized traffic of membranes and membrane-associated proteins. Small GTPases of the Rho family orchestrate cytoskeletal dynamics. Rho GTPase signaling is tightly regulated and mislocalization or constitutive activation may lead to, for example, morphogenetic abnormalities, tumor cell metastasis or apoptosis. There is increasing evidence that traffic to and from the plasma membrane constitutes an important mechanism controlling Rho GTPase activation and signaling. This brief overview discusses a group of proteins that function at the interface between membrane dynamics and RhoGTPase signaling. These proteins all share a so-called BAR domain, which is a lipid and protein binding region that also harbors membrane deforming activity. In the past 15 years, a growing number of BAR domain proteins have been identified and found to regulate Rho GTPase signaling. The studies discussed here define several modes of RhoGTPase regulation through BAR-domain containing proteins, identifying the BAR domain as an important regulatory unit bridging membrane traffic and cytoskeletal dynamics.
Collapse
Affiliation(s)
- Bart-Jan de Kreuk
- Department of Molecular Cell Biology, Sanquin Research and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | | |
Collapse
|
32
|
Razidlo GL, Wang Y, Chen J, Krueger EW, Billadeau DD, McNiven MA. Dynamin 2 potentiates invasive migration of pancreatic tumor cells through stabilization of the Rac1 GEF Vav1. Dev Cell 2013; 24:573-85. [PMID: 23537630 PMCID: PMC3905678 DOI: 10.1016/j.devcel.2013.02.010] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2011] [Revised: 01/03/2013] [Accepted: 02/19/2013] [Indexed: 01/03/2023]
Abstract
The large GTPase Dynamin 2 (Dyn2) is markedly upregulated in pancreatic cancer, is a potent activator of metastatic migration, and is required for Rac1-mediated formation of lamellipodia. Here we demonstrate an unexpected mechanism of Dyn2 action in these contexts via direct binding to the Rac1 guanine nucleotide exchange factor (GEF) Vav1. Surprisingly, disruption of the Dyn2-Vav1 interaction targets Vav1 to the lysosome for degradation via an interaction with the cytoplasmic chaperone Hsc70, resulting in a dramatic reduction of Vav1 protein stability. Importantly, a specific mutation in Vav1 near its Dyn2-binding C-terminal Src homology 3 (SH3) domain prevents Hsc70 binding, resulting in a stabilization of Vav1 levels. Dyn2 binding regulates the interaction of Vav1 with Hsc70 to control the stability and subsequent activity of this oncogenic GEF. These findings elucidate how Dyn2 activates Rac1, lamellipod protrusion, and invasive cellular migration and provide insight into how this specific Vav is ectopically expressed in pancreatic tumors.
Collapse
Affiliation(s)
- Gina L. Razidlo
- Department of Biochemistry and Molecular Biology and the Center for Digestive Diseases, Rochester, Minnesota, 55905 USA
| | - Yu Wang
- Department of Biochemistry and Molecular Biology and the Center for Digestive Diseases, Rochester, Minnesota, 55905 USA
| | - Jing Chen
- Department of Biochemistry and Molecular Biology and the Center for Digestive Diseases, Rochester, Minnesota, 55905 USA
| | - Eugene W. Krueger
- Department of Biochemistry and Molecular Biology and the Center for Digestive Diseases, Rochester, Minnesota, 55905 USA
| | - Daniel D. Billadeau
- Department of Biochemistry and Molecular Biology and the Center for Digestive Diseases, Rochester, Minnesota, 55905 USA
| | - Mark A. McNiven
- Department of Biochemistry and Molecular Biology and the Center for Digestive Diseases, Rochester, Minnesota, 55905 USA
| |
Collapse
|
33
|
Pritz CO, Bitsche M, Salvenmoser W, Dudás J, Schrott-Fischer A, Glueckert R. Endocytic trafficking of silica nanoparticles in a cell line derived from the organ of Corti. Nanomedicine (Lond) 2013; 8:239-52. [DOI: 10.2217/nnm.12.91] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Background: Due to their biochemical versatility, nanoparticles (NPs) have become one of the most important future carriers for drugs and genes. NP-mediated delivery could enable an effective pharmacotherapy to the inner ear and combat hearing loss. Aims: This study investigates the endocytic trafficking of silica NPs within HEI-OC1 cells, a cell line derived from the inner ear. Materials & methods: To investigate the interaction between 50-, 70- and 100-nm silica NPs and the cells, the authors employed a set of commonly available methods involving light and electron microscopy, and sample processing methods, which preserve the native cell shape and the fragile endocytic structures. Results: The authors observed that 50-nm NPs were the most efficiently internalized. They also identified macropinocytosis as the dominant mechanism of uptake, showed localization of NPs in the early endosome and observed that silica NPs were delayed during trafficking to the lysosomes, where these NPs stayed confined, showing no endosomal escape. Conclusion: These silica NPs mostly rely on macropinocytosis for internalization. A successful use of silica NPs as vectors would involve smaller NPs and an endosomal escape strategy. Original submitted 21 December 2011; Revised submitted 23 May 2012; Published online 14 August 2012
Collapse
Affiliation(s)
- Christian Oliver Pritz
- Department of Otolaryngology, Medical University of Innsbruck, 6020 Innsbruck, Anichstraße 35, Austria
| | - Mario Bitsche
- Division of Clinical & Functional Anatomy, Department of Anatomy, Histology & Embryology, Medical University of Innsbruck, Müllerstraße 59, Innsbruck, Austria
| | - Willi Salvenmoser
- Institute of Zoology & Center for Molecular Bioscience, University of Innsbruck, 6020 Innsbruck Technikerstraße 25, Austria
| | - József Dudás
- Department of Otolaryngology, Medical University of Innsbruck, 6020 Innsbruck, Anichstraße 35, Austria
| | - Anneliese Schrott-Fischer
- Department of Otolaryngology, Medical University of Innsbruck, 6020 Innsbruck, Anichstraße 35, Austria
| | - Rudolf Glueckert
- Department of Otolaryngology, Medical University of Innsbruck, 6020 Innsbruck, Anichstraße 35, Austria
- University Clinics of Innsbruck, Tiroler Landeskrankenanstalten GmbH-TILAK, A-6020 Innsbruck, Austria
| |
Collapse
|
34
|
Mu XM, Shi W, Sun LX, Li H, Wang YR, Jiang ZZ, Zhang LY. Pristimerin inhibits breast cancer cell migration by up- regulating regulator of G protein signaling 4 expression. Asian Pac J Cancer Prev 2013; 13:1097-104. [PMID: 22799288 DOI: 10.7314/apjcp.2012.13.4.1097] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND/AIM Pristimerin isolated from Celastrus and Maytenus spp can inhibit proteasome activity. However, whether pristimerin can modulate cancer metastasis is unknown. METHODS The impacts of pristimerin on the purified and intracellular chymotrypsin proteasomal activity, the levels of regulator of G protein signaling 4 (RGS 4) expression and breast cancer cell lamellipodia formation, and the migration and invasion were determined by enzymatic, Western blot, immunofluorescent, and transwell assays, respectively. RESULTS We found that pristimerin inhibited human chymotrypsin proteasomal activity in MDA-MB-231 cells in a dose-dependent manner. Pristimerin also inhibited breast cancer cell lamellipodia formation, migration, and invasion in vitro by up-regulating RGS4 expression. Thus, knockdown of RGS4 attenuated pristimerin-mediated inhibition of breast cancer cell migration and invasion. Furthermore, pristimerin inhibited growth and invasion of implanted breast tumors in mice. CONCLUSION Pristmerin inhibits proteasomal activity and increases the levels of RGS4, inhibiting the migration and invasion of breast cancer cells.
Collapse
Affiliation(s)
- Xian-Min Mu
- Jiangsu Center of Drug Screening, China Pharmaceutical University, Nanjing, China
| | | | | | | | | | | | | |
Collapse
|
35
|
Menon M, Schafer DA. Dynamin: expanding its scope to the cytoskeleton. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2013; 302:187-219. [PMID: 23351711 DOI: 10.1016/b978-0-12-407699-0.00003-0] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The large GTPase dynamin is well known for its actions on budded cellular membranes to generate vesicles, most often, clathrin-coated endocytic vesicles. The scope of cellular processes in which dynamin-mediated vesicle formation occurs, has expanded to include secretory vesicle formation at the Golgi, from other endosomes and nonclathrin structures, such as caveolae, as well as membrane remodeling during exocytosis and vesicle fusion. An intriguing new facet of dynamin's sphere of influence is the cytoskeleton. Cytoskeletal filament networks maintain cell shape, provide cell movement, execute cell division and orchestrate vesicle trafficking. Recent evidence supports the hypothesis that dynamin influences actin filaments and microtubules via mechanisms that are independent of its membrane-remodeling activities. This chapter discusses this emerging evidence and considers possible mechanisms of action.
Collapse
Affiliation(s)
- Manisha Menon
- Department of Biology, University of Virginia, Charlottesville, VA, USA
| | | |
Collapse
|
36
|
Itoh T, Hasegawa J. Mechanistic insights into the regulation of circular dorsal ruffle formation. J Biochem 2012; 153:21-9. [PMID: 23175656 DOI: 10.1093/jb/mvs138] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Growth factor stimulations induce dynamic changes in the cytoskeleton beneath the plasma membrane. Among them is the formation of membrane ruffles organized in a circular array, called 'circular dorsal ruffles' (CDRs). Physiological functions of CDRs include downregulation of cell growth by desensitizing the signalling from growth factor receptors as well as rearrangement of adhesion sites at the onset of cell migration. For the formation of CDRs, not only the activators of actin polymerization, such as N-WASP and the Arp2/3-complex, but also membrane deforming proteins with BAR/F-BAR domains are necessary. Small GTPases are also involved in the formation of CDRs by controlling intracellular trafficking through endosomes. Moreover, recent analyses of another circular cytoskeletal structure, podosome rosettes, have revealed common molecular features shared with CDRs. Among them, the roles of PI3-kinase and phosphoinositide 5-phosphatase may hold the key to the induction of these circular structures.
Collapse
Affiliation(s)
- Toshiki Itoh
- Division of Membrane Biology, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Kobe 650-0017, Japan.
| | | |
Collapse
|
37
|
Mercer J, Helenius A. Gulping rather than sipping: macropinocytosis as a way of virus entry. Curr Opin Microbiol 2012; 15:490-9. [PMID: 22749376 DOI: 10.1016/j.mib.2012.05.016] [Citation(s) in RCA: 158] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2012] [Revised: 05/25/2012] [Accepted: 05/31/2012] [Indexed: 12/18/2022]
Abstract
Macropinocytosis has emerged as a major endocytic mechanism in the cell entry of animal viruses. The process differs fundamentally from other endocytic mechanisms involved in virus internalization. By activating growth factor receptors or other signaling molecules, plasma membrane-bound viruses trigger the activation of a signaling pathway. When amplified, this causes a transient, global change in cell behavior. The consequences of this change include the actin-dependent formation of membrane protrusions, the elevation of non-specific uptake of fluid, and the internalization of membrane together with surface-bound ligands and particles including viruses. Recent studies show that this strategy is used by a variety of enveloped and non-enveloped viruses.
Collapse
Affiliation(s)
- Jason Mercer
- ETH Zürich, Institute of Biochemistry, Zürich, Switzerland.
| | | |
Collapse
|
38
|
African swine fever virus uses macropinocytosis to enter host cells. PLoS Pathog 2012; 8:e1002754. [PMID: 22719252 PMCID: PMC3375293 DOI: 10.1371/journal.ppat.1002754] [Citation(s) in RCA: 149] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2012] [Accepted: 04/30/2012] [Indexed: 12/16/2022] Open
Abstract
African swine fever (ASF) is caused by a large and highly pathogenic DNA virus, African swine fever virus (ASFV), which provokes severe economic losses and expansion threats. Presently, no specific protection or vaccine against ASF is available, despite the high hazard that the continued occurrence of the disease in sub-Saharan Africa, the recent outbreak in the Caucasus in 2007, and the potential dissemination to neighboring countries, represents. Although virus entry is a remarkable target for the development of protection tools, knowledge of the ASFV entry mechanism is still very limited. Whereas early studies have proposed that the virus enters cells through receptor-mediated endocytosis, the specific mechanism used by ASFV remains uncertain. Here we used the ASFV virulent isolate Ba71, adapted to grow in Vero cells (Ba71V), and the virulent strain E70 to demonstrate that entry and internalization of ASFV includes most of the features of macropinocytosis. By a combination of optical and electron microscopy, we show that the virus causes cytoplasm membrane perturbation, blebbing and ruffles. We have also found that internalization of the virions depends on actin reorganization, activity of Na+/H+ exchangers, and signaling events typical of the macropinocytic mechanism of endocytosis. The entry of virus into cells appears to directly stimulate dextran uptake, actin polarization and EGFR, PI3K-Akt, Pak1 and Rac1 activation. Inhibition of these key regulators of macropinocytosis, as well as treatment with the drug EIPA, results in a considerable decrease in ASFV entry and infection. In conclusion, this study identifies for the first time the whole pathway for ASFV entry, including the key cellular factors required for the uptake of the virus and the cell signaling involved. ASFV is a highly pathogenic zoonotic virus, which can cause severe economic losses and bioterrorism threats. No vaccine against ASFV is available so far. A strong hazard of ASFV dissemination through EU countries from Caucasian areas has recently emerged, thus making urgent to acquire knowledge and tools for protection against this virus. Despite that, our understanding of how ASFV enters host cells is very limited. A thorough understanding of this process would enable to design targeted antiviral therapies and vaccine development. The present study clearly defines key steps of ASFV cellular uptake, as well as the host factors responsible for permitting virus entry into cells. Our results indicate that the primary mechanism of ASFV uptake is a macropinocytosis-like process, that involves cellular membrane perturbation, actin polarization, activity of Na+/H+ membrane channels, and signaling proceedings typical of the macropinocytic mechanism of endocytosis, such as Rac1-Pak1 pathways, PI3K and tyrosine-kinases activation. These findings help understanding how ASFV infects cells and suggest that disturbance of macropinocytosis may be useful in the impairment of infection and vaccine development.
Collapse
|
39
|
Feng H, Liu KW, Guo P, Zhang P, Cheng T, McNiven MA, Johnson GR, Hu B, Cheng SY. Dynamin 2 mediates PDGFRα-SHP-2-promoted glioblastoma growth and invasion. Oncogene 2012; 31:2691-702. [PMID: 21996738 PMCID: PMC3262067 DOI: 10.1038/onc.2011.436] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2011] [Revised: 08/17/2011] [Accepted: 08/24/2011] [Indexed: 01/31/2023]
Abstract
Dynamin 2 (Dyn2), a large GTPase, is involved in receptor tyrosine kinase (RTK)-promoted cell migration. However, the molecular mechanisms by which Dyn2 regulates RTK-induced cell migration have not been established. Recently, we reported that tyrosine-protein phosphatase non-receptor type 11 (SHP-2) and phosphatidylinositol 3-kinase (PI3K) mediate platelet-derived growth factor receptor-α (PDGFRα)-promoted glioma tumor growth and invasion. Here, we show that Dyn2 is an effector downstream of the PDGFRα-PI3K/SHP-2 signaling in glioma cells. Depletion of endogenous Dyn2 by short hairpin RNAs (shRNAs) inhibited PDGFRα-stimulated phosphorylation of Akt, Erk1/2, Rac1 and Cdc42 activities, glioma cell migration and survival in vitro and tumor growth and invasion in the brains of mice. Dyn2 binds to SHP-2 and PI3K and colocalizes with PDGFRα at the invasive fronts in PDGF-A-stimulated glioma cells. Inhibition of SHP-2 by siRNA knockdown abrogated Dyn2 association with activated PDGFRα and PDGFRα activation of Rac1 and Cdc42, and glioma cell migration, thereby establishing a link between SHP-2 interaction with Dyn2 and the PDGFRα signaling. Furthermore, a dominant-negative SHP-2 C459S mutant inhibited PDGF-A-stimulated glioma cell migration, phosphorylation of Dyn2 and concomitantly blocked PDGFRα-induced Src activation. Inhibition of Src by Src inhibitors attenuated PDGF-A-stimulated phosphorylation of Akt and Dyn2 and glioma cell migration. Additionally, mutations of binding sites to PI3K, SHP-2 or Src of PDGFRα impaired PDGFRα-stimulated phosphorylation of Akt and Dyn2, and Dyn2 association with activated PDGFRα. Taken together, this study identifies Dyn2 as an effector that mediates PDGFRα-SHP-2-induced glioma tumor growth and invasion, suggesting that targeting the PDGFRα-SHP-2-Dyn2 pathway may be beneficial to patients with malignant glioblastomas.
Collapse
Affiliation(s)
- H Feng
- Cancer Institute University of Pittsburgh, Pittsburgh, PA 15213, USA
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - KW Liu
- Cancer Institute University of Pittsburgh, Pittsburgh, PA 15213, USA
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - P Guo
- Cancer Institute University of Pittsburgh, Pittsburgh, PA 15213, USA
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Division of Pediatric General and Thoracic Surgery, Children's Hospital, Pittsburgh, PA 15201, USA
| | - P Zhang
- Department of Radiation Oncology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - T Cheng
- Department of Radiation Oncology, University of Pittsburgh, Pittsburgh, PA 15213, USA
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Center for Stem Cell Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - MA McNiven
- Department of Biochemistry and Molecular Biology and Center for Basic Research in Digestive Diseases, Mayo Clinic, Rochester, MN 55905, USA
| | - GR Johnson
- Laboratory of Chemistry, Division of Therapeutic Proteins and Center for Drug Evaluation and Research, Food and Drug Administration, Bethesda, MD 20892, USA
| | - B Hu
- Cancer Institute University of Pittsburgh, Pittsburgh, PA 15213, USA
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - SY Cheng
- Cancer Institute University of Pittsburgh, Pittsburgh, PA 15213, USA
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| |
Collapse
|
40
|
Bucci C, Bakke O, Progida C. Charcot-Marie-Tooth disease and intracellular traffic. Prog Neurobiol 2012; 99:191-225. [PMID: 22465036 PMCID: PMC3514635 DOI: 10.1016/j.pneurobio.2012.03.003] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2011] [Revised: 12/23/2011] [Accepted: 03/13/2012] [Indexed: 12/23/2022]
Abstract
Mutations of genes whose primary function is the regulation of membrane traffic are increasingly being identified as the underlying causes of various important human disorders. Intriguingly, mutations in ubiquitously expressed membrane traffic genes often lead to cell type- or organ-specific disorders. This is particularly true for neuronal diseases, identifying the nervous system as the most sensitive tissue to alterations of membrane traffic. Charcot-Marie-Tooth (CMT) disease is one of the most common inherited peripheral neuropathies. It is also known as hereditary motor and sensory neuropathy (HMSN), which comprises a group of disorders specifically affecting peripheral nerves. This peripheral neuropathy, highly heterogeneous both clinically and genetically, is characterized by a slowly progressive degeneration of the muscle of the foot, lower leg, hand and forearm, accompanied by sensory loss in the toes, fingers and limbs. More than 30 genes have been identified as targets of mutations that cause CMT neuropathy. A number of these genes encode proteins directly or indirectly involved in the regulation of intracellular traffic. Indeed, the list of genes linked to CMT disease includes genes important for vesicle formation, phosphoinositide metabolism, lysosomal degradation, mitochondrial fission and fusion, and also genes encoding endosomal and cytoskeletal proteins. This review focuses on the link between intracellular transport and CMT disease, highlighting the molecular mechanisms that underlie the different forms of this peripheral neuropathy and discussing the pathophysiological impact of membrane transport genetic defects as well as possible future ways to counteract these defects.
Collapse
Affiliation(s)
- Cecilia Bucci
- Department of Biological and Environmental Sciences and Technologies (DiSTeBA), University of Salento, Via Provinciale Monteroni, 73100 Lecce, Italy.
| | | | | |
Collapse
|
41
|
Tkachenko E, Tse D, Sideleva O, Deharvengt SJ, Luciano MR, Xu Y, McGarry CL, Chidlow J, Pilch PF, Sessa WC, Toomre DK, Stan RV. Caveolae, fenestrae and transendothelial channels retain PV1 on the surface of endothelial cells. PLoS One 2012; 7:e32655. [PMID: 22403691 PMCID: PMC3293851 DOI: 10.1371/journal.pone.0032655] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2011] [Accepted: 01/28/2012] [Indexed: 11/21/2022] Open
Abstract
PV1 protein is an essential component of stomatal and fenestral diaphragms, which are formed at the plasma membrane of endothelial cells (ECs), on structures such as caveolae, fenestrae and transendothelial channels. Knockout of PV1 in mice results in in utero and perinatal mortality. To be able to interpret the complex PV1 knockout phenotype, it is critical to determine whether the formation of diaphragms is the only cellular role of PV1. We addressed this question by measuring the effect of complete and partial removal of structures capable of forming diaphragms on PV1 protein level. Removal of caveolae in mice by knocking out caveolin-1 or cavin-1 resulted in a dramatic reduction of PV1 protein level in lungs but not kidneys. The magnitude of PV1 reduction correlated with the abundance of structures capable of forming diaphragms in the microvasculature of these organs. The absence of caveolae in the lung ECs did not affect the transcription or translation of PV1, but it caused a sharp increase in PV1 protein internalization rate via a clathrin- and dynamin-independent pathway followed by degradation in lysosomes. Thus, PV1 is retained on the cell surface of ECs by structures capable of forming diaphragms, but undergoes rapid internalization and degradation in the absence of these structures, suggesting that formation of diaphragms is the only role of PV1.
Collapse
Affiliation(s)
- Eugene Tkachenko
- Department of Medicine, University of California, San Diego, California, United States of America
| | - Dan Tse
- Department of Pathology, Dartmouth Medical School, Lebanon, New Hampshire, United States of America
- Heart and Vascular Research Center, Dartmouth Medical School, Lebanon, New Hampshire, United States of America
| | - Olga Sideleva
- Department of Pathology, Dartmouth Medical School, Lebanon, New Hampshire, United States of America
| | - Sophie J. Deharvengt
- Department of Pathology, Dartmouth Medical School, Lebanon, New Hampshire, United States of America
| | - Marcus R. Luciano
- Department of Pathology, Dartmouth Medical School, Lebanon, New Hampshire, United States of America
| | - Yan Xu
- Department of Pathology, Dartmouth Medical School, Lebanon, New Hampshire, United States of America
| | - Caitlin L. McGarry
- Department of Pathology, Dartmouth Medical School, Lebanon, New Hampshire, United States of America
- Heart and Vascular Research Center, Dartmouth Medical School, Lebanon, New Hampshire, United States of America
| | - John Chidlow
- Department of Pharmacology, Yale University, New Haven, Connecticut, United States of America
| | - Paul F. Pilch
- Department of Biochemistry, Boston University, Boston, Massachusetts, United States of America
| | - William C. Sessa
- Department of Pharmacology, Yale University, New Haven, Connecticut, United States of America
| | - Derek K. Toomre
- Department of Cell Biology, Yale University, New Haven, Connecticut, United States of America
| | - Radu V. Stan
- Department of Pathology, Dartmouth Medical School, Lebanon, New Hampshire, United States of America
- Department of Microbiology and Immunology, Dartmouth Medical School, Lebanon, New Hampshire, United States of America
- Heart and Vascular Research Center, Dartmouth Medical School, Lebanon, New Hampshire, United States of America
- Norris Cotton Cancer Center, Dartmouth Medical School, Lebanon, New Hampshire, United States of America
- * E-mail:
| |
Collapse
|
42
|
Ferguson SM, De Camilli P. Dynamin, a membrane-remodelling GTPase. Nat Rev Mol Cell Biol 2012; 13:75-88. [PMID: 22233676 DOI: 10.1038/nrm3266] [Citation(s) in RCA: 751] [Impact Index Per Article: 57.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Dynamin, the founding member of a family of dynamin-like proteins (DLPs) implicated in membrane remodelling, has a critical role in endocytic membrane fission events. The use of complementary approaches, including live-cell imaging, cell-free studies, X-ray crystallography and genetic studies in mice, has greatly advanced our understanding of the mechanisms by which dynamin acts, its essential roles in cell physiology and the specific function of different dynamin isoforms. In addition, several connections between dynamin and human disease have also emerged, highlighting specific contributions of this GTPase to the physiology of different tissues.
Collapse
Affiliation(s)
- Shawn M Ferguson
- Department of Cell Biology, Yale University School of Medicine, New Haven, Connecticut 06510, USA.
| | | |
Collapse
|
43
|
Armstrong SM, Khajoee V, Wang C, Wang T, Tigdi J, Yin J, Kuebler WM, Gillrie M, Davis SP, Ho M, Lee WL. Co-regulation of transcellular and paracellular leak across microvascular endothelium by dynamin and Rac. THE AMERICAN JOURNAL OF PATHOLOGY 2011; 180:1308-1323. [PMID: 22203054 DOI: 10.1016/j.ajpath.2011.12.002] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 05/20/2011] [Revised: 11/23/2011] [Accepted: 12/02/2011] [Indexed: 12/23/2022]
Abstract
Increased permeability of the microvascular endothelium to fluids and proteins is the hallmark of inflammatory conditions such as sepsis. Leakage can occur between (paracellular) or through (transcytosis) endothelial cells, yet little is known about whether these pathways are linked. Understanding the regulation of microvascular permeability is essential for the identification of novel therapies to combat inflammation. We investigated whether transcytosis and paracellular leakage are co-regulated. Using molecular and pharmacologic approaches, we inhibited transcytosis of albumin in primary human microvascular endothelium and measured paracellular permeability. Blockade of transcytosis induced a rapid increase in paracellular leakage that was not explained by decreases in caveolin-1 or increases in activity of nitric oxide synthase. The effect required caveolin-1 but was observed in cells depleted of clathrin, indicating that it was not due to the general inhibition of endocytosis. Inhibiting transcytosis by dynamin blockade increased paracellular leakage concomitantly with the loss of cortical actin from the plasma membrane and the displacement of active Rac from the plasmalemma. Importantly, inhibition of paracellular leakage by sphingosine-1-phosphate, which activates Rac and induces cortical actin, caused a significant increase in transcytosis of albumin in vitro and in an ex vivo whole-lung model. In addition, dominant-negative Rac significantly diminished albumin uptake by endothelia. Our findings indicate that transcytosis and paracellular permeability are co-regulated through a signaling pathway linking dynamin, Rac, and actin.
Collapse
Affiliation(s)
- Susan M Armstrong
- Institute of Medical Science, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Vahid Khajoee
- Keenan Research Centre of the Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario, Canada
| | - Changsen Wang
- Keenan Research Centre of the Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario, Canada
| | - Tieling Wang
- Keenan Research Centre of the Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario, Canada
| | - Jayesh Tigdi
- Keenan Research Centre of the Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario, Canada
| | - Jun Yin
- Keenan Research Centre of the Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario, Canada
| | - Wolfgang M Kuebler
- Keenan Research Centre of the Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario, Canada
| | - Mark Gillrie
- Departments of Microbiology and Infectious Diseases and Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Shevaun P Davis
- Departments of Microbiology and Infectious Diseases and Medicine, University of Calgary, Calgary, Alberta, Canada
| | - May Ho
- Departments of Microbiology and Infectious Diseases and Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Warren L Lee
- Institute of Medical Science, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada; Keenan Research Centre of the Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario, Canada; Division of Respirology and Interdepartmental Division of Critical Care Medicine, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
44
|
Aleksandrowicz P, Marzi A, Biedenkopf N, Beimforde N, Becker S, Hoenen T, Feldmann H, Schnittler HJ. Ebola virus enters host cells by macropinocytosis and clathrin-mediated endocytosis. J Infect Dis 2011; 204 Suppl 3:S957-67. [PMID: 21987776 DOI: 10.1093/infdis/jir326] [Citation(s) in RCA: 185] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Virus entry into host cells is the first step of infection and a crucial determinant of pathogenicity. Here we show that Ebola virus-like particles (EBOV-VLPs) composed of the glycoprotein GP(1,2) and the matrix protein VP40 use macropinocytosis and clathrin-mediated endocytosis to enter cells. EBOV-VLPs applied to host cells induced actin-driven ruffling and enhanced FITC-dextran uptake, which indicated macropinocytosis as the main entry mechanism. This was further supported by inhibition of entry through inhibitors of actin polymerization (latrunculin A), Na(+)/H(+)-exchanger (EIPA), and PI3-kinase (wortmannin). A fraction of EBOV-VLPs, however, colocalized with clathrin heavy chain (CHC), and VLP uptake was reduced by CHC small interfering RNA transfection and expression of the dominant negative dynamin II-K44A mutant. In contrast, we found no evidence that EBOV-VLPs enter cells via caveolae. This work identifies macropinocytosis as the major, and clathrin-dependent endocytosis as an alternative, entry route for EBOV particles. Therefore, EBOV seems to utilize different entry pathways depending on both cell type and virus particle size.
Collapse
Affiliation(s)
- Paulina Aleksandrowicz
- Institute of Anatomy and Vascular Biology, Westfälische Wilhelms University Muenster, Vesaliusweg, Germany
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Antigen stored in dendritic cells after macropinocytosis is released unprocessed from late endosomes to target B cells. Blood 2011; 119:95-105. [PMID: 22049514 DOI: 10.1182/blood-2011-02-336123] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
B lymphocytes can be triggered in lymph nodes by nonopsonized antigens (Ag), potentially in their native form. However, the mechanisms that promote encounter of B lymphocytes with unprocessed antigens in lymph nodes are still elusive. We show here that antigens are detected in B cells in the draining lymph nodes of mice injected with live, but not fixed, dendritic cells (DCs) loaded with antigens. This highlights active processes in DCs to promote Ag transfer to B lymphocytes. In addition, antigen-loaded DCs found in the draining lymph node were CD103+. Using 3 different model Ag, we then show that immature DCs efficiently take up Ag by macropinocytosis and store the internalized material in late endocytic compartments. We find that DCs have a unique ability to release antigens from these compartments in the extracellular medium, which is controlled by Rab27. B cells take up the regurgitated Ag and the chemokine CXCL13, essential to attract B cells in lymph nodes, enhances this transfer. Our results reveal a unique property of DCs to regurgitate unprocessed Ag that could play an important role in B-cell activation.
Collapse
|
46
|
Abstract
Dynamin (Dyn) is a multidomain and multifunctional GTPase best known for its essential role in clathrin-mediated endocytosis (CME). Dyn2 mutations have been linked to two human diseases, centronuclear myopathy (CNM) and Charcot-Marie-Tooth (CMT) disease. Paradoxically, although Dyn2 is ubiquitously expressed and essential for embryonic development, the disease-associated Dyn2 mutants are autosomal dominant, but result in slowly progressing and tissue-specific diseases. Thus, although the cellular defects that cause disease remain unclear, they are expected to be mild. To gain new insight into potential pathogenic mechanisms, we utilized mouse Dyn2 conditional knockout cells combined with retroviral-mediated reconstitution to mimic both heterozygous and homozygous states and characterized cellular phenotypes using quantitative assays for several membrane trafficking events. Surprisingly, none of the four mutants studied exhibited a defect in CME, but all were impaired in their ability to support p75/neurotrophin receptor export from the Golgi, the raft-dependent endocytosis of cholera toxin and the clathrin-independent endocytosis of epidermal growth factor receptor (EGFR). While it will be important to study these mutants in disease-relevant muscle and neuronal cells, given the importance of neurotrophic factors and lipid rafts in muscle physiology, we speculate that these common cellular defects might contribute to the tissue-specific diseases caused by a ubiquitously expressed protein.
Collapse
Affiliation(s)
- Ya-Wen Liu
- Department of Cell Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | | | | |
Collapse
|
47
|
Doherty GJ, Åhlund MK, Howes MT, Morén B, Parton RG, McMahon HT, Lundmark R. The endocytic protein GRAF1 is directed to cell-matrix adhesion sites and regulates cell spreading. Mol Biol Cell 2011; 22:4380-9. [PMID: 21965292 PMCID: PMC3216663 DOI: 10.1091/mbc.e10-12-0936] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
GTPase regulator associated with focal adhesion kinase-1 (GRAF1) interacted with endocytic and adhesion proteins, and GRAF1 endocytic activity was up-regulated in spreading cells and concentrated at the leading edge of migrating cells. Depletion of GRAF1 resulted in profound defects in cell spreading. GRAF1 remodeled membrane microdomains at adhesions, aiding membrane turnover during cell morphological changes. The rho GTPase-activating protein GTPase regulator associated with focal adhesion kinase-1 (GRAF1) remodels membranes into tubulovesicular clathrin-independent carriers (CLICs) mediating lipid-anchored receptor endocytosis. However, the cell biological functions of this highly prevalent endocytic pathway are unclear. In this article, we present biochemical and cell biological evidence that GRAF1 interacted with a network of endocytic and adhesion proteins and was found enriched at podosome-like adhesions and src-induced podosomes. We further demonstrate that these sites comprise microdomains of highly ordered lipid enriched in GRAF1 endocytic cargo. GRAF1 activity was upregulated in spreading cells and uptake via CLICs was concentrated at the leading edge of migrating cells. Depletion of GRAF1, which inhibits CLIC generation, resulted in profound defects in cell spreading and migration. We propose that GRAF1 remodels membrane microdomains at adhesion sites into endocytic carriers, facilitating membrane turnover during cell morphological changes.
Collapse
Affiliation(s)
- Gary J Doherty
- MRC Laboratory of Molecular Biology, Cambridge CB2 0QH, United Kingdom
| | | | | | | | | | | | | |
Collapse
|
48
|
Mulherkar N, Raaben M, de la Torre JC, Whelan SP, Chandran K. The Ebola virus glycoprotein mediates entry via a non-classical dynamin-dependent macropinocytic pathway. Virology 2011; 419:72-83. [PMID: 21907381 DOI: 10.1016/j.virol.2011.08.009] [Citation(s) in RCA: 106] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2011] [Revised: 06/21/2011] [Accepted: 08/08/2011] [Indexed: 12/23/2022]
Abstract
Ebola virus (EBOV) has been reported to enter cultured cell lines via a dynamin-2-independent macropinocytic pathway or clathrin-mediated endocytosis. The route(s) of productive EBOV internalization into physiologically relevant cell types remain unexplored, and viral-host requirements for this process are incompletely understood. Here, we use electron microscopy and complementary chemical and genetic approaches to demonstrate that the viral glycoprotein, GP, induces macropinocytic uptake of viral particles into cells. GP's highly-glycosylated mucin domain is dispensable for virus-induced macropinocytosis, arguing that interactions between other sequences in GP and the host cell surface are responsible. Unexpectedly, we also found a requirement for the large GTPase dynamin-2, which is proposed to be dispensable for several types of macropinocytosis. Our results provide evidence that EBOV uses an atypical dynamin-dependent macropinocytosis-like entry pathway to enter Vero cells, adherent human peripheral blood-derived monocytes, and a mouse dendritic cell line.
Collapse
Affiliation(s)
- Nirupama Mulherkar
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | | | | | | | | |
Collapse
|
49
|
Hasegawa J, Tokuda E, Tenno T, Tsujita K, Sawai H, Hiroaki H, Takenawa T, Itoh T. SH3YL1 regulates dorsal ruffle formation by a novel phosphoinositide-binding domain. ACTA ACUST UNITED AC 2011; 193:901-16. [PMID: 21624956 PMCID: PMC3105542 DOI: 10.1083/jcb.201012161] [Citation(s) in RCA: 73] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Reversible interactions between cytosolic proteins and membrane lipids such as phosphoinositides play important roles in membrane morphogenesis driven by actin polymerization. In this paper, we identify a novel lipid-binding module, which we call the SYLF domain (after the SH3YL1, Ysc84p/Lsb4p, Lsb3p, and plant FYVE proteins that contain it), that is highly conserved from bacteria to mammals. SH3YL1 (SH3 domain containing Ysc84-like 1) strongly bound to phosphatidylinositol 3,4,5-triphosphate (PI(3,4,5)P(3)) and several D5-phosphorylated phosphoinositides through its SYLF domain and was localized to circular dorsal ruffles induced by platelet-derived growth factor stimulation. Interestingly, SHIP2 (the PI(3,4,5)P(3) 5-phosphatase, src-homology 2-containing inositol-5-phosphatase 2) was identified as a binding partner of SH3YL1, and knockdown of these proteins significantly suppressed dorsal ruffle formation. Phosphatidylinositol 3,4-bisphosphate (PI(3,4)P(2)), which is mainly synthesized from PI(3,4,5)P(3) by the action of SHIP2, was enriched in dorsal ruffles, and PI(3,4)P(2) synthesis strongly correlated with formation of the circular membrane structure. These results provide new insight into the molecular mechanism of dorsal ruffle formation and its regulation by phosphoinositide metabolism.
Collapse
Affiliation(s)
- Junya Hasegawa
- Division of Membrane Biology, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe, Hyogo 650-0017, Japan
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Eppinga RD, Krueger EW, Weller SG, Zhang L, Cao H, McNiven MA. Increased expression of the large GTPase dynamin 2 potentiates metastatic migration and invasion of pancreatic ductal carcinoma. Oncogene 2011; 31:1228-41. [PMID: 21841817 PMCID: PMC3218232 DOI: 10.1038/onc.2011.329] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Pancreatic ductal tumors invade local parenchyma and metastasize to distant organs. Src-mediated tyrosine kinase signaling pathways promote pancreatic ductal adenocarcinoma (PDAC) metastasis, though the molecular mechanisms supporting this invasive process are poorly understood and represent important and novel therapeutic targets. The large GTPase Dynamin2 (Dyn2), a Src-kinase substrate, regulates membrane-cytoskeletal dynamics although it is yet to be defined if this mechanoenzyme contributes to tumor cell migration and invasion. Therefore the goal of this study was to test if Dyn2 is upregulated in human pancreatic tumors and to define its role in cell migration and metastatic invasion using in vitro assays and nude mouse models. Histological analysis showed that 81% of the 85 patients tested had elevated Dyn2 in PDAC tissue. To test if Dyn2 overexpression alters metastatic properties of human pancreatic tumor cells, stable clones of BxPC-3 cells overexpressing either wild-type Dyn2 or a phosphorylation-deficient mutant Dyn2Y(231/597)F known to attenuate Dyn2 function, were generated and analyzed for migratory capacity. Importantly, tumor cells expressing 2-3 fold levels of Dyn2 protruded lamellipodia at twice the rate, migrated faster (180%) and farther (2.5-fold greater net distance) on glass and through transwell chambers (2-3 fold more cells through the filter) compared to cells expressing Dyn2Y(231/597)F or vector alone. Further, siRNA-mediated depletion of Dyn2 and dynamin inhibitors MiTMAB and Dynasore significantly reduced cell migration (>66%), wound healing (>75%) and invasion in transwell assays (>95%) compared to DMSO treated cells. To test the metastatic potential conferred by increased Dyn2 expression, the BxPC-3 clonal cell lines were implanted orthotopically into the pancreas of nude mice. Cells expressing Dyn2-GFP exhibited a 3-fold increase in large distal tumors compared to cells expressing Dyn2Y(231/597)F or vector alone. Finally, histological analysis of pancreatic metastases from human patients revealed that Dyn2 is upregulated in 60% of metastatic tumors examined. These findings are the first to implicate dynamin in any neoplastic condition and to directly demonstrate a role for this mechanoenzyme in invasive cell migration.
Collapse
Affiliation(s)
- R D Eppinga
- Department of Biochemistry and Molecular Biology, Center for Digestive Diseases, Rochester, MN, USA
| | | | | | | | | | | |
Collapse
|