1
|
Kumarathasan P, Nazemof N, Blais E, Syama KP, Breznan D, Dirieh Y, Aoki H, Phanse S, Tayabali A, Babu M. In Vitro Exposure of A549 and J774A.1 Cells to SiO 2 and TiO 2 Nanoforms and Related Cellular- and Molecular-Level Effects: Application of Proteomics. J Proteome Res 2025; 24:1672-1687. [PMID: 40036262 PMCID: PMC11976856 DOI: 10.1021/acs.jproteome.4c00651] [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: 07/29/2024] [Revised: 11/22/2024] [Accepted: 01/29/2025] [Indexed: 03/06/2025]
Abstract
There is an emerging interest in incorporating proteomic data for environmental health risk assessments. Meanwhile, the production and use of engineered nanomaterials (ENMs) with attractive physicochemical properties are expanding with the potential for exposure, thus necessitating toxicity information on these materials for health risk analysis, where proteomic data can be informative. Here, cells (A549 human lung epithelial and J774A.1 mouse monocyte/macrophage cells) were exposed to ENMs (nanoforms of SiO2and TiO2) of different sizes and surface chemistries (dose: 0-100 μg/cm2, 24 h) for in vitro toxicity data. Cytotoxicity (CTB, ATP, and LDH), oxidative stress (GSH oxidation), and proteomic analysis (MS- and antibody-based) were conducted post-nanoparticle (NP) exposure to determine the relative potency and identify perturbed cellular pathways. Dose-, nanoform-, and cell type-specific cytotoxicity changes were observed upon exposure to both nanoSiO2 and nanoTiO2. Size, agglomeration, surface modification, and metal impurities appeared to be the determinants of cytotoxicity. Proteomic analysis identified some enriched mechanistic pathways and biological processes relevant to cell defense/phagocytosis, stress, metabolism, apoptosis, and inflammatory processes in J774A.1 cells exposed to these NPs. A549 cells exhibited enriched pathway/biological processes relevant to transport/endocytosis, stress, metabolism, and inflammatory processes post-NP exposures. Concordance was observed between the nanoform exposure- and cell type-related cytotoxicity responses, notably cellular ATP, which is critical for cell viability, oxidative stress, and cellular pathways/biological processes. These findings demonstrate the application of proteomics in regulatory toxicology and warrant further research in this direction.
Collapse
Affiliation(s)
- Premkumari Kumarathasan
- Environmental
Health Science and Research Bureau, HECSB, Health Canada, Ottawa, Ontario, Canada K1A 0K9
- Faculty
of Health Sciences, University of Ottawa, Ottawa, Ontario, Canada K1N 6N5
| | - Nazila Nazemof
- Environmental
Health Science and Research Bureau, HECSB, Health Canada, Ottawa, Ontario, Canada K1A 0K9
- Faculty
of Health Sciences, University of Ottawa, Ottawa, Ontario, Canada K1N 6N5
| | - Erica Blais
- Environmental
Health Science and Research Bureau, HECSB, Health Canada, Ottawa, Ontario, Canada K1A 0K9
| | - Krishna Priya Syama
- Environmental
Health Science and Research Bureau, HECSB, Health Canada, Ottawa, Ontario, Canada K1A 0K9
| | - Dalibor Breznan
- Environmental
Health Science and Research Bureau, HECSB, Health Canada, Ottawa, Ontario, Canada K1A 0K9
| | - Yasmine Dirieh
- Environmental
Health Science and Research Bureau, HECSB, Health Canada, Ottawa, Ontario, Canada K1A 0K9
| | - Hiroyuki Aoki
- Department
of Biochemistry, University of Regina, Regina, Saskatchewan, Canada S4S 0A2
| | - Sadhna Phanse
- Department
of Biochemistry, University of Regina, Regina, Saskatchewan, Canada S4S 0A2
| | - Azam Tayabali
- Environmental
Health Science and Research Bureau, HECSB, Health Canada, Ottawa, Ontario, Canada K1A 0K9
| | - Mohan Babu
- Department
of Biochemistry, University of Regina, Regina, Saskatchewan, Canada S4S 0A2
| |
Collapse
|
2
|
Galvão GF, Petrilli R, Arfelli VC, Carvalho AN, Martins YA, Rosales RRC, Archangelo LF, daSilva LLP, Lopez RFV. Iontophoresis-driven alterations in nanoparticle uptake pathway and intracellular trafficking in carcinoma skin cancer cells. Colloids Surf B Biointerfaces 2025; 248:114459. [PMID: 39709939 DOI: 10.1016/j.colsurfb.2024.114459] [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: 08/28/2024] [Revised: 12/13/2024] [Accepted: 12/15/2024] [Indexed: 12/24/2024]
Abstract
Effective treatment of squamous cell carcinoma (SCC) poses challenges due to intrinsic drug resistance and limited drug penetration into tumor cells. Nanoparticle-based drug delivery systems have emerged as a promising approach to enhance therapeutic efficacy; however, they often face hurdles such as inadequate cellular uptake and rapid lysosomal degradation. This study explores the potential of iontophoresis to augment the efficacy of liposome and immunoliposome-based drug delivery systems for SCC treatment. The study assessed iontophoresis effects on SCC cell line (A431) viability, nanoparticle uptake dynamics, and intracellular distribution patterns. Specific inhibitors were employed to delineate cellular internalization pathways, while fluorescence microscopy and immunohistochemistry examined changes in EGFR expression and lysosomal activity. Results demonstrated that iontophoresis significantly increased cellular uptake of liposomes and immunoliposomes, achieving approximately 50 % uptake compared to 10 % with passive treatment. This enhancement correlated with modifications in endocytic pathways, favoring macropinocytosis and caveolin-mediated endocytosis for liposomes, and macropinocytosis and clathrin-mediated pathways for immunoliposomes. Moreover, iontophoresis induced alterations in EGFR distribution and triggered syncytium-like cellular clustering. It also attenuated lysosomal activity, thereby reducing nanoparticle degradation and prolonging intracellular retention of therapeutic agents. These findings underscore the role of iontophoresis in modulating nanoparticle internalization pathways, offering insights that could advance targeted drug delivery strategies and mitigate therapeutic resistance in SCC and other malignancies.
Collapse
Affiliation(s)
- Gabriela Fávero Galvão
- School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP 14020-630, Brazil
| | - Raquel Petrilli
- School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP 14020-630, Brazil; Institute of Health Sciences, University for International Integration of the Afro-Brazilian Lusophony, Redenção, CE, Brazil; Federal University of Ceara, Faculty of Pharmacy, Dentistry and Nursing, Department of Pharmacy, Fortaleza, Ceará, Brazil
| | - Vanessa Cristina Arfelli
- Department of Cell and Molecular Biology and Pathogenic Bioagents, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Andréia Nogueira Carvalho
- Department of Cell and Molecular Biology and Pathogenic Bioagents, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Yugo Araújo Martins
- School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP 14020-630, Brazil
| | - Roberta Ribeiro Costa Rosales
- Department of Cell and Molecular Biology and Pathogenic Bioagents, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Leticia Fröhlich Archangelo
- Department of Cell and Molecular Biology and Pathogenic Bioagents, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Luis Lamberti Pinto daSilva
- Department of Cell and Molecular Biology and Pathogenic Bioagents, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Renata Fonseca Vianna Lopez
- School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP 14020-630, Brazil.
| |
Collapse
|
3
|
Thayer RC, Polston ES, Xu J, Begun DJ. Regional specialization, polyploidy, and seminal fluid transcripts in the Drosophila female reproductive tract. Proc Natl Acad Sci U S A 2024; 121:e2409850121. [PMID: 39453739 PMCID: PMC11536144 DOI: 10.1073/pnas.2409850121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 09/20/2024] [Indexed: 10/27/2024] Open
Abstract
Sexual reproduction requires the choreographed interaction of female cells and molecules with sperm and seminal fluid. In internally fertilizing animals, these interactions are managed by specialized tissues within the female reproductive tract (FRT), such as a uterus, glands, and sperm storage organs. However, female somatic reproductive tissues remain understudied, hindering insight into the molecular interactions that support fertility. Here, we report the identification, molecular characterization, and analysis of cell types throughout the somatic FRT in the premier Drosophila melanogaster model system. We find that the uterine epithelia is composed of 11 distinct cell types with well-delineated spatial domains, likely corresponding to functionally specialized surfaces that interact with gametes and reproductive fluids. Polyploidy is pervasive: More than half of lower reproductive tract cells are ≥4C. While seminal fluid proteins (SFPs) are typically thought of as male products that are transferred to females, we find that specialized cell types in the sperm storage organs heavily invest in expressing SFP genes. Rates of amino acid divergence between closely related species indicate heterogeneous evolutionary processes acting on male-limited versus female-expressed seminal fluid genes. Together, our results emphasize that more than 40% of annotated seminal fluid genes are better described as shared components of reproductive transcriptomes, which may function cooperatively to support spermatozoa. More broadly, our work provides the molecular foundation for improved technologies to catalyze the functional characterization of the FRT.
Collapse
Affiliation(s)
- Rachel C. Thayer
- Department of Evolution and Ecology, University of California, Davis, CA95616
| | | | - Jixiang Xu
- Department of Evolution and Ecology, University of California, Davis, CA95616
| | - David J. Begun
- Department of Evolution and Ecology, University of California, Davis, CA95616
| |
Collapse
|
4
|
Carvalho Cabral P, Richard VR, Borchers CH, Olivier M, Cermakian N. Circadian Control of the Response of Macrophages to Plasmodium Spp.-Infected Red Blood Cells. Immunohorizons 2024; 8:442-456. [PMID: 38916585 PMCID: PMC11220744 DOI: 10.4049/immunohorizons.2400021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 05/23/2024] [Indexed: 06/26/2024] Open
Abstract
Malaria is a serious vector-borne disease characterized by periodic episodes of high fever and strong immune responses that are coordinated with the daily synchronized parasite replication cycle inside RBCs. As immune cells harbor an autonomous circadian clock that controls various aspects of the immune response, we sought to determine whether the intensity of the immune response to Plasmodium spp., the parasite causing malaria, depends on time of infection. To do this, we developed a culture model in which mouse bone marrow-derived macrophages are stimulated with RBCs infected with Plasmodium berghei ANKA (iRBCs). Lysed iRBCs, but not intact iRBCs or uninfected RBCs, triggered an inflammatory immune response in bone marrow-derived macrophages. By stimulating at four different circadian time points (16, 22, 28, or 34 h postsynchronization of the cells' clock), 24-h rhythms in reactive oxygen species and cytokines/chemokines were found. Furthermore, the analysis of the macrophage proteome and phosphoproteome revealed global changes in response to iRBCs that varied according to circadian time. This included many proteins and signaling pathways known to be involved in the response to Plasmodium infection. In summary, our findings show that the circadian clock within macrophages determines the magnitude of the inflammatory response upon stimulation with ruptured iRBCs, along with changes of the cell proteome and phosphoproteome.
Collapse
Affiliation(s)
| | - Vincent R. Richard
- Lady Davis Institute for Medical Research, McGill University, Montreal, Quebec, Canada
| | - Christoph H. Borchers
- Lady Davis Institute for Medical Research, McGill University, Montreal, Quebec, Canada
| | - Martin Olivier
- Research Institute of the McGill University Health Centre, McGill University, Montreal, Quebec, Canada
| | - Nicolas Cermakian
- Douglas Research Centre, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
5
|
Koganti PP, Zhao AH, Selvaraj V. Exogenous cholesterol acquisition signaling in LH-responsive MA-10 Leydig cells and in adult mice. J Endocrinol 2022; 254:187-199. [PMID: 35900012 PMCID: PMC9840751 DOI: 10.1530/joe-22-0043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 07/27/2022] [Indexed: 01/17/2023]
Abstract
MA-10 cells, established 4 decades ago from a murine Leydig cell tumor, has served as a key model system for studying steroidogenesis. Despite a precipitous loss in their innate ability to respond to luteinizing hormone (LH), the use of a cell-permeable cAMP analog for induction ensured their continued use. In parallel, a paradigm that serum-free conditions are essential for trophic steroidogenic stimulation was rationalized. Through the selection of LH-responsive single-cell MA-10Slip clones, we uncovered that Leydig cells remain responsive in the presence of serum in vitro and that exogenous cholesterol delivery by lipoproteins provided a significantly elevated steroid biosynthetic response (>2-fold). In scrutinizing the underlying regulation, systems biology of the MA-10 cell proteome identified multiple Rho-GTPase signaling pathways as highly enriched. Testing Rho function in steroidogenesis revealed that its modulation can negate the specific elevation in steroid biosynthesis observed in the presence of lipoproteins/serum. This signaling modality primarily linked to the regulation of endocytic traffic is evident only in the presence of exogenous cholesterol. Inhibiting Rho function in vivo also decreased hCG-induced testosterone production in mice. Collectively, our findings dispel a long-held view that the use of serum could confound or interfere with trophic stimulation and underscore the need for exogenous lipoproteins when dissecting physiological signaling and cholesterol trafficking for steroid biosynthesis in vitro. The LH-responsive MA-10Slip clones derived in this study present a reformed platform enabling biomimicry to study the cellular and molecular basis of mammalian steroidogenesis.
Collapse
Affiliation(s)
- Prasanthi P. Koganti
- Department of Animal Science, College of Agriculture and Life Sciences, Cornell University, Ithaca, NY 14853, USA
| | - Amy H. Zhao
- Department of Animal Science, College of Agriculture and Life Sciences, Cornell University, Ithaca, NY 14853, USA
| | - Vimal Selvaraj
- Department of Animal Science, College of Agriculture and Life Sciences, Cornell University, Ithaca, NY 14853, USA
- Correspondence should be addressed to: Vimal Selvaraj, Department of Animal Science, College of Agriculture and Life Sciences, Cornell University, Ithaca, NY 14853; ; Tel. 607-255-6138; Fax. 607-255-9829
| |
Collapse
|
6
|
Wan B, Poirié M, Gatti JL. Parasitoid wasp venom vesicles (venosomes) enter Drosophila melanogaster lamellocytes through a flotillin/lipid raft-dependent endocytic pathway. Virulence 2020; 11:1512-1521. [PMID: 33135553 PMCID: PMC7605353 DOI: 10.1080/21505594.2020.1838116] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 10/12/2020] [Accepted: 10/14/2020] [Indexed: 12/22/2022] Open
Abstract
Venosomes are extracellular vesicles found in the venom of Leptopilina endoparasitoids wasps, which transport and target virulence factors to impair the parasitoid egg encapsulation by the lamellocytes of their Drosophila melanogaster host larva. Using the co-immunolocalization of fluorescent L. boulardi venosomes and one of the putative-transported virulence factors, LbGAP, with known markers of cellular endocytosis, we show that venosomes endocytosis by lamellocytes is not a process dependent on clathrin or macropinocytosis and internalization seems to bypass the early endosomal compartment Rab5. After internalization, LbGAP colocalizes strongly with flotillin-1 and the GPI-anchored protein Atilla/L1 (a lamellocyte surface marker) suggesting that entry occurs via a flotillin/lipid raft-dependent pathway. Once internalized, venosomes reach all intracellular compartments, including late and recycling endosomes, lysosomes, and the endoplasmic reticulum network. Venosomes therefore enter their target cells by a specific mechanism and the virulence factors are widely distributed in the lamellocytes' compartments to impair their functions.
Collapse
Affiliation(s)
- Bin Wan
- Université Côte d’Azur, INRAE, CNRS, ISA, France
| | | | | |
Collapse
|
7
|
Jeger JL. Endosomes, lysosomes, and the role of endosomal and lysosomal biogenesis in cancer development. Mol Biol Rep 2020; 47:9801-9810. [PMID: 33185829 DOI: 10.1007/s11033-020-05993-4] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 11/02/2020] [Accepted: 11/06/2020] [Indexed: 12/19/2022]
Abstract
Endosomes and lysosomes are membrane-bound organelles crucial for the normal functioning of the eukaryotic cell. The primary function of endosomes relates to the transportation of extracellular material into the intracellular domain. Lysosomes, on the other hand, are primarily involved in the degradation of macromolecules. Endosomes and lysosomes interact through two distinct pathways: kiss-and-run and direct fusion. In addition to the internalization of particles, endosomes also play an important role in cell signaling and autophagy. Disruptions in either of these processes may contribute to cancer development. Lysosomal proteins, such as cathepsins, can play a role in both tumorigenesis and cancer cell apoptosis. Since endosomal and lysosomal biogenesis and signaling are important components of normal cellular growth and proliferation, proteins involved in these processes are attractive targets for cancer research and, potentially, therapeutics. This literature review provides an overview of the endocytic pathway, endolysosome formation, and the interplay between endosomal/lysosomal biogenesis and carcinogenesis.
Collapse
|
8
|
Wesén E, Lundmark R, Esbjörner EK. Role of Membrane Tension Sensitive Endocytosis and Rho GTPases in the Uptake of the Alzheimer's Disease Peptide Aβ(1-42). ACS Chem Neurosci 2020; 11:1925-1936. [PMID: 32497421 PMCID: PMC7497631 DOI: 10.1021/acschemneuro.0c00053] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Intraneuronal accumulation of amyloid-β (Aβ) is an early pathological signum of Alzheimer's disease, and compartments of the endolysosomal system have been implicated in both seeding and cell-cell propagation of Aβ aggregation. We have studied how clathrin-independent mechanisms contribute to Aβ endocytosis, exploring pathways that are sensitive to changes in membrane tension and the regulation of Rho GTPases. Using live cell confocal microscopy and flow cytometry, we show the uptake of monomeric Aβ(1-42) into endocytic vesicles and vacuole-like dilations, following relaxation of osmotic pressure-induced cell membrane tension. This indicates Aβ(1-42) uptake via clathrin independent carriers (CLICs), although overexpression of the bar-domain protein GRAF1, a key regulator of CLICs, had no apparent effect. We furthermore report reduced Aβ(1-42) uptake following overexpression of constitutively active forms of the Rho GTPases Cdc42 and RhoA, whereas modulation of Rac1, which is linked to macropinosome formation, had no effect. Our results confirm that uptake of Aβ(1-42) is clathrin- and dynamin-independent and point to the involvement of a new and distinct clathrin-independent endocytic mechanism which is similar to uptake via CLICs or macropinocytosis but that also appear to involve yet uncharacterized molecular players.
Collapse
Affiliation(s)
- Emelie Wesén
- Division of Chemical Biology, Department of Biology and Biological Engineering, Chalmers University of Technology, Kemivägen 10, 412 96 Gothenburg, Sweden
| | - Richard Lundmark
- Department of Integrative Medical Biology, Umeå University, Umeå 901 87, Sweden
| | - Elin K. Esbjörner
- Division of Chemical Biology, Department of Biology and Biological Engineering, Chalmers University of Technology, Kemivägen 10, 412 96 Gothenburg, Sweden
| |
Collapse
|
9
|
Hasan M, Khatun A, Fukuta T, Kogure K. Noninvasive transdermal delivery of liposomes by weak electric current. Adv Drug Deliv Rev 2020; 154-155:227-235. [PMID: 32589904 DOI: 10.1016/j.addr.2020.06.016] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 05/27/2020] [Accepted: 06/16/2020] [Indexed: 02/06/2023]
Abstract
Noninvasive transdermal drug delivery (NTDD) offers an exciting new method of administration relative to conventional routes, but is associated with some challenges. Liposomes are capable of encapsulating transdermally-unfavorable drugs. However, the horny layer of skin is a significant barrier that limits efficient transdermal delivery of liposomes. Iontophoresis using weak electric current (WEC) represents a NTDD technology. WEC treatment of liposomes applied to the skin surface improves transdermal penetration of encapsulated drugs by cooperative effects. In this review, we provide an overview of the application of WEC/liposomes for transdermal delivery of macromolecules and low molecular weight drugs. We compare the transdermal delivery and therapeutic efficiency of the combined system with conventional routes of administration and their individual use. We discuss a novel perspective on the mechanism of WEC-mediated transdermal delivery of liposomes, which suggests that WEC activates the intracellular signaling pathway for transdermal permeation and induces unique endocytosis in skin cells.
Collapse
Affiliation(s)
- Mahadi Hasan
- Department of Pharmaceutical Health Chemistry, Tokushima University Graduate School of Biomedical Sciences, Tokushima 770-8505, Japan; Tokyo Biochemical Research Foundation (TBRF) Fellow, Tokushima, Japan
| | - Anowara Khatun
- Department of Pharmaceutical Health Chemistry, Tokushima University Graduate School of Biomedical Sciences, Tokushima 770-8505, Japan
| | - Tatsuya Fukuta
- Department of Pharmaceutical Health Chemistry, Tokushima University Graduate School of Biomedical Sciences, Tokushima 770-8505, Japan
| | - Kentaro Kogure
- Department of Pharmaceutical Health Chemistry, Tokushima University Graduate School of Biomedical Sciences, Tokushima 770-8505, Japan.
| |
Collapse
|
10
|
Tang K, Li S, Li P, Xia Q, Yang R, Li T, Li L, Jiang Y, Qin X, Yang H, Wu C, You F, Tan Y, Liu Y. Shear stress stimulates integrin β1 trafficking and increases directional migration of cancer cells via promoting deacetylation of microtubules. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2020; 1867:118676. [DOI: 10.1016/j.bbamcr.2020.118676] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/08/2019] [Revised: 01/09/2020] [Accepted: 02/05/2020] [Indexed: 12/17/2022]
|
11
|
Nipah Virus-Like Particle Egress Is Modulated by Cytoskeletal and Vesicular Trafficking Pathways: a Validated Particle Proteomics Analysis. mSystems 2019; 4:4/5/e00194-19. [PMID: 31551400 PMCID: PMC6759566 DOI: 10.1128/msystems.00194-19] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Classified as a biosafety level 4 (BSL4) select agent, Nipah virus (NiV) is a deadly henipavirus in the Paramyxoviridae family, with a nearly 75% mortality rate in humans, underscoring its global and animal health importance. Elucidating the process of viral particle production in host cells is imperative both for targeted drug design and viral particle-based vaccine development. However, little is understood concerning the functions of cellular machinery in paramyxoviral and henipaviral assembly and budding. Recent studies showed evidence for the involvement of multiple NiV proteins in viral particle formation, in contrast to the mechanisms understood for several paramyxoviruses as being reliant on the matrix (M) protein alone. Further, the levels and purposes of cellular factor incorporation into viral particles are largely unexplored for the paramyxoviruses. To better understand the involvement of cellular machinery and the major structural viral fusion (F), attachment (G), and matrix (M) proteins, we performed proteomics analyses on virus-like particles (VLPs) produced from several combinations of these NiV proteins. Our findings indicate that NiV VLPs incorporate vesicular trafficking and actin cytoskeletal factors. The involvement of these biological processes was validated by experiments indicating that the perturbation of key factors in these cellular processes substantially modulated viral particle formation. These effects were most impacted for NiV-F-modulated viral particle formation either autonomously or in combination with other NiV proteins, indicating that NiV-F budding relies heavily on these cellular processes. These findings indicate a significant involvement of the NiV fusion protein, vesicular trafficking, and actin cytoskeletal processes in efficient viral particle formation.IMPORTANCE Nipah virus is a zoonotic biosafety level 4 agent with high mortality rates in humans. The genus to which Nipah virus belongs, Henipavirus, includes five officially recognized pathogens; however, over 20 species have been identified in multiple continents within the last several years. As there are still no vaccines or treatments for NiV infection, elucidating its process of viral particle production is imperative both for targeted drug design as well as for particle-based vaccine development. Developments in high-throughput technologies make proteomic analysis of isolated viral particles a highly insightful approach to understanding the life cycle of pathogens such as Nipah virus.
Collapse
|
12
|
Strain-Dependent Porcine Circovirus Type 2 (PCV2) Entry and Replication in T-Lymphoblasts. Viruses 2019; 11:v11090813. [PMID: 31480752 PMCID: PMC6783876 DOI: 10.3390/v11090813] [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: 08/05/2019] [Revised: 08/29/2019] [Accepted: 08/31/2019] [Indexed: 12/25/2022] Open
Abstract
Porcine circovirus type 2 (PCV2) is the etiological agent of PCV2-associated diseases (PCVAD). PCV2 targets lymphoblasts, and pigs suffering from PCVAD display lymphocyte depletion in lymphoid tissues. PCV2 infection of lymphoblasts has not been studied. Here, the replication cycle of PCV2 (abortion strain 1121 and PMWS strain Stoon1010) in T-lymphoblasts was examined. The expression of Rep and Cap were found for both viral strains, while progeny virus was detected for Stoon1010 but not for 1121. PCV2 attached to 11–26% (1121-Stoon1010) of the T-lymphoblasts while 2.6–12.7% of cells showed virus internalization. Chondroitin sulfate (CS) was present on 25% of T-lymphoblasts, and colocalized with PCV2 on 31–32% of the PCV2+ cells. Enzymatic removal of CS reduced PCV2 infection. PCV2 infection was decreased by chlorpromazine, cytochalasin D and Clostridium difficile toxin B for both viral strains and by amiloride for 1121 but not for Stoon1010. Inhibiting either endosome acidification or serine proteases strongly reduced PCV2 infection. Three-dimensional analysis of Cap structure demonstrated a better Cap-nucleic acid affinity for Stoon1010 than for 1121. Taken together, PCV2 binds to T-lymphoblasts partially via CS, enters via clathrin-mediated endocytosis, and disassembles under functions of a pH-drop and serine proteases. Strain Stoon1010 displayed an enhanced viral binding, a specific receptor-mediated endocytosis, an increased Cap-nucleic acid affinity, and a more productive infection in T-lymphoblasts than 1121 did, indicating an evolution from 1121 to Stoon1010.
Collapse
|
13
|
Baratchi S, Keov P, Darby WG, Lai A, Khoshmanesh K, Thurgood P, Vahidi P, Ejendal K, McIntyre P. The TRPV4 Agonist GSK1016790A Regulates the Membrane Expression of TRPV4 Channels. Front Pharmacol 2019; 10:6. [PMID: 30728775 PMCID: PMC6351496 DOI: 10.3389/fphar.2019.00006] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Accepted: 01/04/2019] [Indexed: 02/05/2023] Open
Abstract
TRPV4 is a non-selective cation channel that tunes the function of different tissues including the vascular endothelium, lung, chondrocytes, and neurons. GSK1016790A is the selective and potent agonist of TRPV4 and a pharmacological tool that is used to study the TRPV4 physiological function in vitro and in vivo. It remains unknown how the sensitivity of TRPV4 to this agonist is regulated. The spatial and temporal dynamics of receptors are the major determinants of cellular responses to stimuli. Membrane translocation has been shown to control the response of several members of the transient receptor potential (TRP) family of ion channels to different stimuli. Here, we show that TRPV4 stimulation with GSK1016790A caused an increase in [Ca2+]i that is stable for a few minutes. Single molecule analysis of TRPV4 channels showed that the density of TRPV4 at the plasma membrane is controlled through two modes of membrane trafficking, complete, and partial vesicular fusion. Further, we show that the density of TRPV4 at the plasma membrane decreased within 20 min, as they translocate to the recycling endosomes and that the surface density is dependent on the release of calcium from the intracellular stores and is controlled via a PI3K, PKC, and RhoA signaling pathway.
Collapse
Affiliation(s)
- Sara Baratchi
- School of Health and Biomedical Sciences, RMIT University, Melbourne, VIC, Australia
| | - Peter Keov
- School of Health and Biomedical Sciences, RMIT University, Melbourne, VIC, Australia.,Molecular Pharmacology Division, Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia.,St Vincent's Clinical School, University of New South Wales, Darlinghurst, NSW, Australia
| | - William G Darby
- School of Health and Biomedical Sciences, RMIT University, Melbourne, VIC, Australia
| | - Austin Lai
- School of Health and Biomedical Sciences, RMIT University, Melbourne, VIC, Australia
| | | | - Peter Thurgood
- School of Engineering, RMIT University, Melbourne, VIC, Australia
| | - Parisa Vahidi
- School of Health and Biomedical Sciences, RMIT University, Melbourne, VIC, Australia
| | - Karin Ejendal
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, United States
| | - Peter McIntyre
- School of Health and Biomedical Sciences, RMIT University, Melbourne, VIC, Australia
| |
Collapse
|
14
|
Xia Y, Xie Y, Yu Z, Xiao H, Jiang G, Zhou X, Yang Y, Li X, Zhao M, Li L, Zheng M, Han S, Zong Z, Meng X, Deng H, Ye H, Fa Y, Wu H, Oldfield E, Hu X, Liu W, Shi Y, Zhang Y. The Mevalonate Pathway Is a Druggable Target for Vaccine Adjuvant Discovery. Cell 2018; 175:1059-1073.e21. [PMID: 30270039 DOI: 10.1016/j.cell.2018.08.070] [Citation(s) in RCA: 165] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Revised: 08/13/2018] [Accepted: 08/30/2018] [Indexed: 01/02/2023]
Abstract
Motivated by the clinical observation that interruption of the mevalonate pathway stimulates immune responses, we hypothesized that this pathway may function as a druggable target for vaccine adjuvant discovery. We found that lipophilic statin drugs and rationally designed bisphosphonates that target three distinct enzymes in the mevalonate pathway have potent adjuvant activities in mice and cynomolgus monkeys. These inhibitors function independently of conventional "danger sensing." Instead, they inhibit the geranylgeranylation of small GTPases, including Rab5 in antigen-presenting cells, resulting in arrested endosomal maturation, prolonged antigen retention, enhanced antigen presentation, and T cell activation. Additionally, inhibiting the mevalonate pathway enhances antigen-specific anti-tumor immunity, inducing both Th1 and cytolytic T cell responses. As demonstrated in multiple mouse cancer models, the mevalonate pathway inhibitors are robust for cancer vaccinations and synergize with anti-PD-1 antibodies. Our research thus defines the mevalonate pathway as a druggable target for vaccine adjuvants and cancer immunotherapies.
Collapse
Affiliation(s)
- Yun Xia
- School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology, Tsinghua University, 100084 Beijing, China; Joint Graduate Program of Peking-Tsinghua-NIBS, School of Life Sciences, Tsinghua University, 100084 Beijing, China
| | - Yonghua Xie
- School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology, Tsinghua University, 100084 Beijing, China
| | - Zhengsen Yu
- School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology, Tsinghua University, 100084 Beijing, China
| | - Hongying Xiao
- School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology, Tsinghua University, 100084 Beijing, China; Joint Graduate Program of Peking-Tsinghua-NIBS, School of Life Sciences, Tsinghua University, 100084 Beijing, China; Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu, 610041 Sichuan, China
| | - Guimei Jiang
- School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology, Tsinghua University, 100084 Beijing, China; Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu, 610041 Sichuan, China
| | - Xiaoying Zhou
- School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology, Tsinghua University, 100084 Beijing, China
| | - Yunyun Yang
- School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology, Tsinghua University, 100084 Beijing, China
| | - Xin Li
- School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology, Tsinghua University, 100084 Beijing, China; Joint Graduate Program of Peking-Tsinghua-NIBS, School of Life Sciences, Tsinghua University, 100084 Beijing, China
| | - Meng Zhao
- Joint Graduate Program of Peking-Tsinghua-NIBS, School of Life Sciences, Tsinghua University, 100084 Beijing, China; MOE Key Laboratory of Protein Sciences, School of Life Sciences, Tsinghua University, 100084 Beijing, China
| | - Liping Li
- School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology, Tsinghua University, 100084 Beijing, China
| | - Mingke Zheng
- Institute for Immunology and School of Medicine, Tsinghua University, 100084 Beijing, China
| | - Shuai Han
- School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology, Tsinghua University, 100084 Beijing, China
| | - Zhaoyun Zong
- MOE Key Laboratory of Bioinformatics, School of Life Sciences, Tsinghua University, 100084 Beijing, China
| | - Xianbin Meng
- MOE Key Laboratory of Bioinformatics, School of Life Sciences, Tsinghua University, 100084 Beijing, China
| | - Haiteng Deng
- MOE Key Laboratory of Bioinformatics, School of Life Sciences, Tsinghua University, 100084 Beijing, China
| | - Huahu Ye
- Laboratory Animal Center, Academy of Military Medical Sciences, 100071 Beijing, China
| | - Yunzhi Fa
- Laboratory Animal Center, Academy of Military Medical Sciences, 100071 Beijing, China
| | - Haitao Wu
- Department of Neurobiology, Beijing Institute of Basic Medical Sciences, Academy of Military Medical Sciences, 100850 Beijing, China
| | - Eric Oldfield
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Xiaoyu Hu
- Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu, 610041 Sichuan, China; Institute for Immunology and School of Medicine, Tsinghua University, 100084 Beijing, China
| | - Wanli Liu
- MOE Key Laboratory of Protein Sciences, School of Life Sciences, Tsinghua University, 100084 Beijing, China; Institute for Immunology and School of Medicine, Tsinghua University, 100084 Beijing, China.
| | - Yan Shi
- Institute for Immunology and School of Medicine, Tsinghua University, 100084 Beijing, China; Institute Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute, University of Calgary, Calgary, AB, Canada.
| | - Yonghui Zhang
- School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology, Tsinghua University, 100084 Beijing, China; Joint Graduate Program of Peking-Tsinghua-NIBS, School of Life Sciences, Tsinghua University, 100084 Beijing, China; Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu, 610041 Sichuan, China.
| |
Collapse
|
15
|
Zhang Y, Wang QC, Liu J, Xiong B, Cui XS, Kim NH, Sun SC. The small GTPase CDC42 regulates actin dynamics during porcine oocyte maturation. J Reprod Dev 2017; 63:505-510. [PMID: 28781348 PMCID: PMC5649100 DOI: 10.1262/jrd.2017-034] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The mammalian oocyte undergoes an asymmetric division during meiotic maturation, producing a small polar body and a haploid gamete. This process involves the dynamics of actin filaments, and the guanosine triphosphatase (GTPase) protein superfamily is a major regulator of actin assembly. In the present study, the small GTPase CDC42 was shown to participate in the meiotic maturation of porcine oocytes. Immunofluorescent staining showed that CDC42 was mainly localized at the periphery of the oocytes, and accumulated with microtubules. Deactivation of CDC42 protein activity with the effective inhibitor ML141 caused a decrease in actin distribution in the cortex, which resulted in a failure of polar body extrusion. Moreover, western blot analysis revealed that besides the Cdc42-N-WASP pathway previously reported in mouse oocytes, the expression of ROCK and p-cofilin, two molecules involved in actin dynamics, was also decreased after CDC42 inhibition during porcine oocyte maturation. Thus, our study demonstrates that CDC42 is an indispensable protein during porcine oocyte meiosis, and CDC42 may interact with N-WASP, ROCK, and cofilin in the assembly of actin filaments during porcine oocyte maturation.
Collapse
Affiliation(s)
- Yu Zhang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Qiao-Chu Wang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Jun Liu
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Bo Xiong
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Xiang-Shun Cui
- Department of Animal Sciences, Chungbuk National University, Cheongju 361-763, Korea
| | - Nam-Hyung Kim
- Department of Animal Sciences, Chungbuk National University, Cheongju 361-763, Korea
| | - Shao-Chen Sun
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| |
Collapse
|
16
|
Schulz AM, Stutte S, Hogl S, Luckashenak N, Dudziak D, Leroy C, Forné I, Imhof A, Müller SA, Brakebusch CH, Lichtenthaler SF, Brocker T. Cdc42-dependent actin dynamics controls maturation and secretory activity of dendritic cells. J Cell Biol 2016; 211:553-67. [PMID: 26553928 PMCID: PMC4639873 DOI: 10.1083/jcb.201503128] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Cdc42 control of actin dynamics keeps DCs in an immature state, and loss of Cdc42 activity facilitates secretion and rapid up-regulation of intracellular molecules to the cell surface, which shows that Cdc42 contributes to DC immunogenicity by regulating the DC actin cytoskeleton. Cell division cycle 42 (Cdc42) is a member of the Rho guanosine triphosphatase family and has pivotal functions in actin organization, cell migration, and proliferation. To further study the molecular mechanisms of dendritic cell (DC) regulation by Cdc42, we used Cdc42-deficient DCs. Cdc42 deficiency renders DCs phenotypically mature as they up-regulate the co-stimulatory molecule CD86 from intracellular storages to the cell surface. Cdc42 knockout DCs also accumulate high amounts of invariant chain–major histocompatibility complex (MHC) class II complexes at the cell surface, which cannot efficiently present peptide antigens (Ag’s) for priming of Ag-specific CD4 T cells. Proteome analyses showed a significant reduction in lysosomal MHC class II–processing proteins, such as cathepsins, which are lost from DCs by enhanced secretion. As these effects on DCs can be mimicked by chemical actin disruption, our results propose that Cdc42 control of actin dynamics keeps DCs in an immature state, and cessation of Cdc42 activity during DC maturation facilitates secretion as well as rapid up-regulation of intracellular molecules to the cell surface.
Collapse
Affiliation(s)
- Anna M Schulz
- Institute for Immunology, Ludwig Maximilians University Munich, 80336 Munich, Germany
| | - Susanne Stutte
- Institute for Immunology, Ludwig Maximilians University Munich, 80336 Munich, Germany
| | - Sebastian Hogl
- Deutsches Zentrum für Neurodegenerative Erkrankungen, 81377 Munich, Germany
| | - Nancy Luckashenak
- Institute for Immunology, Ludwig Maximilians University Munich, 80336 Munich, Germany
| | - Diana Dudziak
- Department of Dermatology, University Hospital of Erlangen, 91052 Erlangen, Germany
| | - Céline Leroy
- Institute for Immunology, Ludwig Maximilians University Munich, 80336 Munich, Germany
| | - Ignasi Forné
- Adolf Butenandt Institute, Ludwig Maximilians University Munich, 80336 Munich, Germany
| | - Axel Imhof
- Adolf Butenandt Institute, Ludwig Maximilians University Munich, 80336 Munich, Germany
| | - Stephan A Müller
- Deutsches Zentrum für Neurodegenerative Erkrankungen, 81377 Munich, Germany
| | - Cord H Brakebusch
- Molecular Pathology Section, Biotech Research and Innovation Center, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Stefan F Lichtenthaler
- Munich Cluster for Systems Neurology, Ludwig Maximilians University Munich, 80336 Munich, Germany Deutsches Zentrum für Neurodegenerative Erkrankungen, 81377 Munich, Germany Neuroproteomics, Klinikum rechts der Isar, Institute for Advanced Study, Technische Universität München, 80333 Munich, Germany
| | - Thomas Brocker
- Institute for Immunology, Ludwig Maximilians University Munich, 80336 Munich, Germany
| |
Collapse
|
17
|
Salinas RP, Ortiz Flores RM, Distel JS, Aguilera MO, Colombo MI, Berón W. Coxiella burnetii Phagocytosis Is Regulated by GTPases of the Rho Family and the RhoA Effectors mDia1 and ROCK. PLoS One 2015; 10:e0145211. [PMID: 26674774 PMCID: PMC4682630 DOI: 10.1371/journal.pone.0145211] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Accepted: 12/01/2015] [Indexed: 01/09/2023] Open
Abstract
The GTPases belonging to the Rho family control the actin cytoskeleton rearrangements needed for particle internalization during phagocytosis. ROCK and mDia1 are downstream effectors of RhoA, a GTPase involved in that process. Coxiella burnetii, the etiologic agent of Q fever, is internalized by the host´s cells in an actin-dependent manner. Nevertheless, the molecular mechanism involved in this process has been poorly characterized. This work analyzes the role of different GTPases of the Rho family and some downstream effectors in the internalization of C. burnetii by phagocytic and non-phagocytic cells. The internalization of C. burnetii into HeLa and RAW cells was significantly inhibited when the cells were treated with Clostridium difficile Toxin B which irreversibly inactivates members of the Rho family. In addition, the internalization was reduced in HeLa cells that overexpressed the dominant negative mutants of RhoA, Rac1 or Cdc42 or that were knocked down for the Rho GTPases. The pharmacological inhibition or the knocking down of ROCK diminished bacterium internalization. Moreover, C. burnetii was less efficiently internalized in HeLa cells overexpressing mDia1-N1, a dominant negative mutant of mDia1, while the overexpression of the constitutively active mutant mDia1-ΔN3 increased bacteria uptake. Interestingly, when HeLa and RAW cells were infected, RhoA, Rac1 and mDia1 were recruited to membrane cell fractions. Our results suggest that the GTPases of the Rho family play an important role in C. burnetii phagocytosis in both HeLa and RAW cells. Additionally, we present evidence that ROCK and mDia1, which are downstream effectors of RhoA, are involved in that process.
Collapse
Affiliation(s)
- Romina P. Salinas
- Instituto de Histología y Embriología, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo—CONICET, Mendoza, 5500, Argentina
| | - Rodolfo M. Ortiz Flores
- Instituto de Histología y Embriología, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo—CONICET, Mendoza, 5500, Argentina
| | - Jesús S. Distel
- Instituto de Histología y Embriología, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo—CONICET, Mendoza, 5500, Argentina
| | - Milton O. Aguilera
- Instituto de Histología y Embriología, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo—CONICET, Mendoza, 5500, Argentina
| | - María I. Colombo
- Instituto de Histología y Embriología, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo—CONICET, Mendoza, 5500, Argentina
| | - Walter Berón
- Instituto de Histología y Embriología, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo—CONICET, Mendoza, 5500, Argentina
- * E-mail:
| |
Collapse
|
18
|
Amphetamine activates Rho GTPase signaling to mediate dopamine transporter internalization and acute behavioral effects of amphetamine. Proc Natl Acad Sci U S A 2015; 112:E7138-47. [PMID: 26553986 DOI: 10.1073/pnas.1511670112] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Acute amphetamine (AMPH) exposure elevates extracellular dopamine through a variety of mechanisms that include inhibition of dopamine reuptake, depletion of vesicular stores, and facilitation of dopamine efflux across the plasma membrane. Recent work has shown that the DAT substrate AMPH, unlike cocaine and other nontransported blockers, can also stimulate endocytosis of the plasma membrane dopamine transporter (DAT). Here, we show that when AMPH enters the cytoplasm it rapidly stimulates DAT internalization through a dynamin-dependent, clathrin-independent process. This effect, which can be observed in transfected cells, cultured dopamine neurons, and midbrain slices, is mediated by activation of the small GTPase RhoA. Inhibition of RhoA activity with C3 exotoxin or a dominant-negative RhoA blocks AMPH-induced DAT internalization. These actions depend on AMPH entry into the cell and are blocked by the DAT inhibitor cocaine. AMPH also stimulates cAMP accumulation and PKA-dependent inactivation of RhoA, thus providing a mechanism whereby PKA- and RhoA-dependent signaling pathways can interact to regulate the timing and robustness of AMPH's effects on DAT internalization. Consistent with this model, the activation of D1/D5 receptors that couple to PKA in dopamine neurons antagonizes RhoA activation, DAT internalization, and hyperlocomotion observed in mice after AMPH treatment. These observations support the existence of an unanticipated intracellular target that mediates the effects of AMPH on RhoA and cAMP signaling and suggest new pathways to target to disrupt AMPH action.
Collapse
|
19
|
Kortüm F, Harms FL, Hennighausen N, Rosenberger G. αPIX Is a Trafficking Regulator that Balances Recycling and Degradation of the Epidermal Growth Factor Receptor. PLoS One 2015; 10:e0132737. [PMID: 26177020 PMCID: PMC4503440 DOI: 10.1371/journal.pone.0132737] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2014] [Accepted: 06/17/2015] [Indexed: 12/14/2022] Open
Abstract
Endosomal sorting is an essential control mechanism for signaling through the epidermal growth factor receptor (EGFR). We report here that the guanine nucleotide exchange factor αPIX, which modulates the activity of Rho-GTPases, is a potent bimodal regulator of EGFR trafficking. αPIX interacts with the E3 ubiquitin ligase c-Cbl, an enzyme that attaches ubiquitin to EGFR, thereby labelling this tyrosine kinase receptor for lysosomal degradation. We show that EGF stimulation induces αPIX::c-Cbl complex formation. Simultaneously, αPIX and c-Cbl protein levels decrease, which depends on both αPIX binding to c-Cbl and c-Cbl ubiquitin ligase activity. Through interaction αPIX sequesters c-Cbl from EGFR and this results in reduced EGFR ubiquitination and decreased EGFR degradation upon EGF treatment. However, quantitatively more decisive for cellular EGFR distribution than impaired EGFR degradation is a strong stimulating effect of αPIX on EGFR recycling to the cell surface. This function depends on the GIT binding domain of αPIX but not on interaction with c-Cbl or αPIX exchange activity. In summary, our data demonstrate a previously unappreciated function of αPIX as a strong promoter of EGFR recycling. We suggest that the novel recycling regulator αPIX and the degradation factor c-Cbl closely cooperate in the regulation of EGFR trafficking: uncomplexed αPIX and c-Cbl mediate a positive and a negative feedback on EGFR signaling, respectively; αPIX::c-Cbl complex formation, however, results in mutual inhibition, which may reflect a stable condition in the homeostasis of EGF-induced signal flow.
Collapse
Affiliation(s)
- Fanny Kortüm
- Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Frederike Leonie Harms
- Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Natascha Hennighausen
- Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Georg Rosenberger
- Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- * E-mail:
| |
Collapse
|
20
|
Braun AC, Hendrick J, Eisler SA, Schmid S, Hausser A, Olayioye MA. The Rho-specific GAP protein DLC3 coordinates endocytic membrane trafficking. J Cell Sci 2015; 128:1386-99. [PMID: 25673874 DOI: 10.1242/jcs.163857] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Membrane trafficking is known to be coordinated by small GTPases, but the identity of their regulators, the guanine nucleotide exchange factors (GEFs) and GTPase-activating proteins (GAPs) that ensure balanced GTPase activation at different subcellular sites is largely elusive. Here, we show in living cells that deleted in liver cancer 3 (DLC3, also known as STARD8) is a functional Rho-specific GAP protein, the loss of which enhances perinuclear RhoA activity. DLC3 is recruited to Rab8-positive membrane tubules and is required for the integrity of the Rab8 and Golgi compartments. Depletion of DLC3 impairs the transport of internalized transferrin to the endocytic recycling compartment (ERC), which is restored by the simultaneous downregulation of RhoA and RhoB. We further demonstrate that DLC3 loss interferes with epidermal growth factor receptor (EGFR) degradation associated with prolonged receptor signaling. Taken together, these findings identify DLC3 as a novel component of the endocytic trafficking machinery, wherein it maintains organelle integrity and regulates membrane transport through the control of Rho activity.
Collapse
Affiliation(s)
- Anja C Braun
- Institute of Cell Biology and Immunology, University of Stuttgart, Allmandring 31, 70569 Stuttgart, Germany
| | - Janina Hendrick
- Institute of Cell Biology and Immunology, University of Stuttgart, Allmandring 31, 70569 Stuttgart, Germany
| | - Stephan A Eisler
- Institute of Cell Biology and Immunology, University of Stuttgart, Allmandring 31, 70569 Stuttgart, Germany
| | - Simone Schmid
- Institute of Cell Biology and Immunology, University of Stuttgart, Allmandring 31, 70569 Stuttgart, Germany
| | - Angelika Hausser
- Institute of Cell Biology and Immunology, University of Stuttgart, Allmandring 31, 70569 Stuttgart, Germany
| | - Monilola A Olayioye
- Institute of Cell Biology and Immunology, University of Stuttgart, Allmandring 31, 70569 Stuttgart, Germany
| |
Collapse
|
21
|
Azzarelli R, Guillemot F, Pacary E. Function and regulation of Rnd proteins in cortical projection neuron migration. Front Neurosci 2015; 9:19. [PMID: 25705175 PMCID: PMC4319381 DOI: 10.3389/fnins.2015.00019] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2014] [Accepted: 01/13/2015] [Indexed: 01/08/2023] Open
Abstract
The mammalian cerebral cortex contains a high variety of neuronal subtypes that acquire precise spatial locations and form long or short-range connections to establish functional neuronal circuits. During embryonic development, cortical projection neurons are generated in the areas lining the lateral ventricles and they subsequently undergo radial migration to reach the position of their final maturation within the cortical plate. The control of the neuroblast migratory behavior and the coordination of the migration process with other neurogenic events such as cell cycle exit, differentiation and final maturation are crucial to normal brain development. Among the key regulators of cortical neuron migration, the small GTP binding proteins of the Rho family and the atypical Rnd members play important roles in integrating intracellular signaling pathways into changes in cytoskeletal dynamics and motility behavior. Here we review the role of Rnd proteins during cortical neuronal migration and we discuss both the upstream mechanisms that regulate Rnd protein activity and the downstream molecular pathways that mediate Rnd effects on cell cytoskeleton.
Collapse
Affiliation(s)
- Roberta Azzarelli
- Cambridge Department of Oncology, Hutchison/MRC Research Centre, University of Cambridge Cambridge, UK
| | - François Guillemot
- Division of Molecular Neurobiology, MRC National Institute for Medical Research London, UK
| | - Emilie Pacary
- Institut National de la Santé et de la Recherche Médicale U862, Neurocentre Magendie Bordeaux, France ; Université de Bordeaux Bordeaux, France
| |
Collapse
|
22
|
Yang Z, Sun D, Yan Z, Reynolds AB, Christman JW, Minshall RD, Malik AB, Zhang Y, Hu G. Differential role for p120-catenin in regulation of TLR4 signaling in macrophages. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2014; 193:1931-1941. [PMID: 25015829 PMCID: PMC4119481 DOI: 10.4049/jimmunol.1302863] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Activation of TLR signaling through recognition of pathogen-associated molecular patterns is essential for the innate immune response against bacterial and viral infections. We have shown that p120-catenin (p120) suppresses TLR4-mediated NF-кB signaling in LPS-challenged endothelial cells. In this article, we report that p120 differentially regulates LPS/TLR4 signaling in mouse bone marrow-derived macrophages. We observed that p120 inhibited MyD88-dependent NF-κB activation and release of TNF-α and IL-6, but enhanced TIR domain-containing adapter-inducing IFN-β-dependent IFN regulatory factor 3 activation and release of IFN-β upon LPS exposure. p120 silencing diminished LPS-induced TLR4 internalization, whereas genetic and pharmacological inhibition of RhoA GTPase rescued the decrease in endocytosis of TLR4 and TLR4-MyD88 signaling, and reversed the increase in TLR4-TIR domain-containing adapter-inducing IFN-β signaling induced by p120 depletion. Furthermore, we demonstrated that altered p120 expression in macrophages regulates the inflammatory phenotype of LPS-induced acute lung injury. These results indicate that p120 functions as a differential regulator of TLR4 signaling pathways by facilitating TLR4 endocytic trafficking in macrophages, and support a novel role for p120 in influencing the macrophages in the lung inflammatory response to endotoxin.
Collapse
Affiliation(s)
- Zhiyong Yang
- Department of Anesthesiology, University of Illinois College of Medicine, Chicago, IL 60612
| | - Dong Sun
- Department of Anesthesiology, University of Illinois College of Medicine, Chicago, IL 60612; Department of General Surgery, Qilu Hospital of Shandong University, Jinan, 250012 China
| | - Zhibo Yan
- Department of Anesthesiology, University of Illinois College of Medicine, Chicago, IL 60612; Department of General Surgery, Qilu Hospital of Shandong University, Jinan, 250012 China
| | - Albert B Reynolds
- Department of Cancer Biology, Vanderbilt University, Nashville, TN 37232
| | - John W Christman
- Department of Medicine, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210; and
| | - Richard D Minshall
- Department of Anesthesiology, University of Illinois College of Medicine, Chicago, IL 60612; Department of Pharmacology, University of Illinois College of Medicine, Chicago, IL 60612
| | - Asrar B Malik
- Department of Pharmacology, University of Illinois College of Medicine, Chicago, IL 60612
| | - Yang Zhang
- Department of Anesthesiology, University of Illinois College of Medicine, Chicago, IL 60612
| | - Guochang Hu
- Department of Anesthesiology, University of Illinois College of Medicine, Chicago, IL 60612; Department of Pharmacology, University of Illinois College of Medicine, Chicago, IL 60612
| |
Collapse
|
23
|
Thompson DB, Villaseñor R, Dorr BM, Zerial M, Liu DR. Cellular uptake mechanisms and endosomal trafficking of supercharged proteins. ACTA ACUST UNITED AC 2014; 19:831-43. [PMID: 22840771 DOI: 10.1016/j.chembiol.2012.06.014] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2012] [Revised: 05/17/2012] [Accepted: 06/21/2012] [Indexed: 02/08/2023]
Abstract
Supercharged proteins (SCPs) can deliver functional macromolecules into the cytoplasm of mammalian cells more potently than unstructured cationic peptides. Thus far, neither the structural features of SCPs that determine their delivery effectiveness nor their intracellular fate postendocytosis, has been studied. Using a large set of supercharged GFP (scGFP) variants, we found that the level of cellular uptake is sigmoidally related to net charge and that scGFPs enter cells through multiple pathways, including clathrin-dependent endocytosis and macropinocytosis. SCPs activate Rho and ERK1/2 and also alter the endocytosis of transferrin and EGF. Finally, we discovered that the intracellular trafficking of endosomes containing scGFPs is altered in a manner that correlates with protein delivery potency. Collectively, our findings establish basic structure-activity relationships of SCPs and implicate the modulation of endosomal trafficking as a determinant of macromolecule delivery efficiency.
Collapse
Affiliation(s)
- David B Thompson
- Howard Hughes Medical Institute, Department of Chemistry and Chemical Biology, Harvard University, 12 Oxford Street, Cambridge, MA 02138, USA
| | | | | | | | | |
Collapse
|
24
|
Grossman GH, Ebke LA, Beight CD, Jang GF, Crabb JW, Hagstrom SA. Protein partners of dynamin-1 in the retina. Vis Neurosci 2013; 30:129-39. [PMID: 23746204 PMCID: PMC3936680 DOI: 10.1017/s0952523813000138] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Dynamin proteins are involved in vesicle generation, providing mechanical force to excise newly formed vesicles from membranes of cellular compartments. In the brain, dynamin-1, dynamin-2, and dynamin-3 have been well studied; however, their function in the retina remains elusive. A retina-specific splice variant of dynamin-1 interacts with the photoreceptor-specific protein Tubby-like protein 1 (Tulp1), which when mutated causes an early onset form of autosomal recessive retinitis pigmentosa. Here, we investigated the role of the dynamins in the retina, using immunohistochemistry to localize dynamin-1, dynamin-2, and dynamin-3 and immunoprecipitation followed by mass spectrometry to explore dynamin-1 interacting proteins in mouse retina. Dynamin-2 is primarily confined to the inner segment compartment of photoreceptors, suggesting a role in outer segment protein transport. Dynamin-3 is present in the terminals of photoreceptors and dendrites of second-order neurons but is most pronounced in the inner plexiform layer where second-order neurons relay signals from photoreceptors. Dynamin-1 appears to be the dominant isoform in the retina and is present throughout the retina and in multiple compartments of the photoreceptor cell. This suggests that it may function in multiple cellular pathways. Surprisingly, dynamin-1 expression and localization did not appear to be disrupted in tulp1−/− mice. Immunoprecipitation experiments reveal that dynamin-1 associates primarily with proteins involved in cytoskeletal-based membrane dynamics. This finding is confirmed by western blot analysis. Results further implicate dynamin-1 in vesicular protein transport processes relevant to synaptic and post-Golgi pathways and indicate a possible role in photoreceptor stability.
Collapse
Affiliation(s)
- Gregory H Grossman
- Department of Ophthalmic Research, Cleveland Clinic Cole Eye Institute, Cleveland, Ohio
| | | | | | | | | | | |
Collapse
|
25
|
Pathak R, Dermardirossian C. GEF-H1: orchestrating the interplay between cytoskeleton and vesicle trafficking. Small GTPases 2013; 4:174-9. [PMID: 23648943 DOI: 10.4161/sgtp.24616] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Vesicle trafficking is crucial for delivery of membrane compartments as well as signaling molecules to specific sites on the plasma membrane for regulation of diverse processes such as cell division, migration, polarity establishment and secretion. Rho GTPases are well-studied signaling molecules that regulate actin cytoskeleton in response to variety of extracellular stimuli. Increasing amounts of evidence suggest that Rho proteins play a critical role in vesicle trafficking in both the exocytic and endocytic pathways; however, the molecular mechanism underlying the process remains largely unclear. We recently defined a mechanism of action for RhoA in membrane trafficking pathways through regulation of the octameric complex exocyst in a manuscript published in Developmental Cell. We have shown that microtubule-associated RhoA-activating factor GEF-H1 is involved in endocytic and excocytic vesicle trafficking. GEF-H1 activates RhoA in response to RalA GTPase, which in turn regulates the localization and the assembly of exocyst components and exocytosis. Our work defines a mechanism for RhoA activation in response to RalA signaling and during vesicle trafficking. These results provide a framework for understanding how RhoA/GEF-H1 regulates the coordination of actin and microtubule cytoskeleton modulation and vesicle trafficking during migration and cell division.
Collapse
Affiliation(s)
- Ritu Pathak
- Departments of Immunology and Microbial Science; The Scripps Research Institute; La Jolla, CA USA
| | - Celine Dermardirossian
- Departments of Immunology and Microbial Science; The Scripps Research Institute; La Jolla, CA USA
| |
Collapse
|
26
|
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
|
27
|
Jux B, Staratschek-Jox A, Penninger JM, Schultze JL, Kolanus W. Vav1 regulates MHCII expression in murine resting and activated B cells. Int Immunol 2013; 25:307-17. [PMID: 23391492 DOI: 10.1093/intimm/dxs157] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Vav1 is a guanine nucleotide exchange factor (GEF) for Rho GTPases, which is exclusively expressed in cells of the hematopoietic system. In addition to its well-documented GEF activity, it was suggested to have other functions due to the presence of multiple domains and nuclear localization signals in its protein structure. Although GEF-dependent and GEF-independent functions of vav have been implicated in T-cell development and T-cell receptor signaling, the role of vav1 in antigen-presenting cells is poorly understood. We found that vav1 is an important regulator of MHCII expression and transport. Microarray analysis of unstimulated bone marrow-derived macrophages revealed a novel role of vav1 in transcriptional regulation of the MHCII locus, possibly by indirect means. Primary immune cells from vav1-deficient mice had a significantly lower constitutive surface expression of MHCII with the strongest impact observed on splenic and peritoneal B cells. Impaired MHCII expression resulted in a diminished capacity for T-cell activation. Using 6-thio-GTP, a specific inhibitor of the GEF function of vav1, we were able to show that the GEF activity is required for MHCII upregulation in B cells after stimulation with LPS. Furthermore, our data show that vav1 not only affects transcription of the MHCII locus but also is an important regulator of MHCII protein transport to the cell surface.
Collapse
Affiliation(s)
- Bettina Jux
- Department of Molecular Immune and Cell Biology, Life and Medical Sciences (LIMES) Institute, University of Bonn, Germany.
| | | | | | | | | |
Collapse
|
28
|
Luo R, Akpan IO, Hayashi R, Sramko M, Barr V, Shiba Y, Randazzo PA. GTP-binding protein-like domain of AGAP1 is protein binding site that allosterically regulates ArfGAP protein catalytic activity. J Biol Chem 2012; 287:17176-17185. [PMID: 22453919 DOI: 10.1074/jbc.m111.334458] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
AGAPs are a subtype of Arf GTPase-activating proteins (GAPs) with 11 members in humans. In addition to the Arf GAP domain, the proteins contain a G-protein-like domain (GLD) with homology to Ras superfamily proteins and a PH domain. AGAPs bind to clathrin adaptors, function in post Golgi membrane traffic, and have been implicated in glioblastoma. The regulation of AGAPs is largely unexplored. Other enzymes containing GTP binding domains are regulated by nucleotide binding. However, nucleotide binding to AGAPs has not been detected. Here, we found that neither nucleotides nor deleting the GLD of AGAP1 affected catalysis, which led us to hypothesize that the GLD is a protein binding site that regulates GAP activity. Two-hybrid screens identified RhoA, Rac1, and Cdc42 as potential binding partners. Coimmunoprecipitation confirmed that AGAP1 and AGAP2 can bind to RhoA. Binding was mediated by the C terminus of RhoA and was independent of nucleotide. RhoA and the C-terminal peptide from RhoA increased GAP activity specifically for the substrate Arf1. In contrast, a C-terminal peptide from Cdc42 neither bound nor activated AGAP1. Based on these results, we propose that AGAPs are allosterically regulated through protein binding to the GLD domain.
Collapse
Affiliation(s)
- Ruibai Luo
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland 20892
| | - Itoro O Akpan
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland 20892
| | - Ryo Hayashi
- Department of Chemistry, Faculty of Science and Engineering, Saga University, Honjo, Saga 840-8502, Japan
| | - Marek Sramko
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland 20892
| | - Valarie Barr
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland 20892
| | - Yoko Shiba
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland 20892
| | - Paul A Randazzo
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland 20892.
| |
Collapse
|
29
|
Illuminating the functional and structural repertoire of human TBC/RABGAPs. Nat Rev Mol Cell Biol 2012; 13:67-73. [PMID: 22251903 DOI: 10.1038/nrm3267] [Citation(s) in RCA: 124] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The Tre2-Bub2-Cdc16 (TBC) domain-containing RAB-specific GTPase-activating proteins (TBC/RABGAPs) are characterized by the presence of highly conserved TBC domains and act as negative regulators of RABs. The importance of TBC/RABGAPs in the regulation of specific intracellular trafficking routes is now emerging, as is their role in different diseases. Importantly, TBC/RABGAPs act as key regulatory nodes, integrating signalling between RABs and other small GTPases and ensuring the appropriate retrieval, transport and delivery of different intracellular vesicles.
Collapse
|
30
|
Small rho GTPases and cholesterol biosynthetic pathway intermediates in African swine fever virus infection. J Virol 2011; 86:1758-67. [PMID: 22114329 DOI: 10.1128/jvi.05666-11] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The integrity of the cholesterol biosynthesis pathway is required for efficient African swine fever virus (ASFV) infection. Incorporation of prenyl groups into Rho GTPases plays a key role in several stages of ASFV infection, since both geranylgeranyl and farnesyl pyrophosphates are required at different infection steps. We found that Rho GTPase inhibition impaired virus morphogenesis and resulted in an abnormal viral factory size with the accumulation of envelope precursors and immature virions. Furthermore, abundant defective virions reached the plasma membrane, and filopodia formation in exocytosis was abrogated. Rac1 was activated at early ASFV infection stages, coincident with microtubule acetylation, a process that stabilizes microtubules for virus transport. Rac1 inhibition did not affect the viral entry step itself but impaired subsequent virus production. We found that specific Rac1 inhibition impaired viral induced microtubule acetylation and viral intracellular transport. These findings highlight that viral infection is the result of a carefully orchestrated modulation of Rho family GTPase activity within the host cell; this modulation results critical for virus morphogenesis and in turn, triggers cytoskeleton remodeling, such as microtubule stabilization for viral transport during early infection.
Collapse
|
31
|
Fazlollahi F, Sipos A, Kim YH, Hamm-Alvarez SF, Borok Z, Kim KJ, Crandall ED. Translocation of PEGylated quantum dots across rat alveolar epithelial cell monolayers. Int J Nanomedicine 2011; 6:2849-57. [PMID: 22131830 PMCID: PMC3224711 DOI: 10.2147/ijn.s26051] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND In this study, primary rat alveolar epithelial cell monolayers (RAECM) were used to investigate transalveolar epithelial quantum dot trafficking rates and underlying transport mechanisms. METHODS Trafficking rates of quantum dots (PEGylated CdSe/ZnS, core size 5.3 nm, hydrodynamic size 25 nm) in the apical-to-basolateral direction across RAECM were determined. Changes in bioelectric properties (ie, transmonolayer resistance and equivalent active ion transport rate) of RAECM in the presence or absence of quantum dots were measured. Involvement of endocytic pathways in quantum dot trafficking across RAECM was assessed using specific inhibitors (eg, methyl-β-cyclodextrin, chlorpromazine, and dynasore for caveolin-, clathrin-, and dynamin-mediated endocytosis, respectively). The effects of lowering tight junctional resistance on quantum dot trafficking were determined by depleting Ca(2+) in apical and basolateral bathing fluids of RAECM using 2 mM EGTA. Effects of temperature on quantum dot trafficking were studied by lowering temperature from 37°C to 4°C. RESULTS Apical exposure of RAECM to quantum dots did not elicit changes in transmonolayer resistance or ion transport rate for up to 24 hours; quantum dot trafficking rates were not surface charge-dependent; methyl-β-cyclodextrin, chlorpromazine, and dynasore did not decrease quantum dot trafficking rates; lowering of temperature decreased transmonolayer resistance by approximately 90% with a concomitant increase in quantum dot trafficking by about 80%; and 24 hours of treatment of RAECM with EGTA decreased transmonolayer resistance by about 95%, with increased quantum dot trafficking of up to approximately 130%. CONCLUSION These data indicate that quantum dots do not injure RAECM and that quantum dot trafficking does not appear to take place via endocytic pathways involving caveolin, clathrin, or dynamin. We conclude that quantum dot translocation across RAECM takes place via both transcellular and paracellular pathways and, based on comparison with our prior studies, interactions of nanoparticles with RAECM are strongly dependent on nanoparticle composition and surface properties.
Collapse
Affiliation(s)
- Farnoosh Fazlollahi
- Will Rogers Institute Pulmonary Research Center, University of Southern California, Los Angeles, CA, USA
| | | | | | | | | | | | | |
Collapse
|
32
|
Huang M, Satchell L, DuHadaway JB, Prendergast GC, Laury-Kleintop LD. RhoB links PDGF signaling to cell migration by coordinating activation and localization of Cdc42 and Rac. J Cell Biochem 2011; 112:1572-84. [PMID: 21344485 PMCID: PMC3079809 DOI: 10.1002/jcb.23069] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The small GTPase RhoB regulates endocytic trafficking of receptor tyrosine kinases (RTKs) and the non-receptor kinases Src and Akt. While receptor-mediated endocytosis is critical for signaling processes driving cell migration, mechanisms that coordinate endocytosis with the propagation of migratory signals remain relatively poorly understood. In this study, we show that RhoB is essential for activation and trafficking of the key migratory effectors Cdc42 and Rac in mediating the ability of platelet-derived growth factor (PDGF) to stimulate cell movement. Stimulation of the PDGF receptor-β on primary vascular smooth muscle cells (VSMCs) results in RhoB-dependent trafficking of endosome-bound Cdc42 from the perinuclear region to the cell periphery, where the RhoGEF Vav2 and Rac are also recruited to drive formation of circular dorsal and peripheral ruffles necessary for cell migration. Our findings identify a novel RhoB-dependent endosomal trafficking pathway that integrates RTK endocytosis with Cdc42/Rac localization and cell movement.
Collapse
Affiliation(s)
- Minzhou Huang
- Lankenau Institute for Medical Research, Wynnewood PA USA
| | | | | | - George C. Prendergast
- Lankenau Institute for Medical Research, Wynnewood PA USA
- Department of Pathology, Anatomy and Cell Biology and Kimmel Cancer Center, Thomas Jefferson University, Philadelphia PA USA
| | | |
Collapse
|
33
|
Raposo RAS, Thomas B, Ridlova G, James W. Proteomic-based identification of CD4-interacting proteins in human primary macrophages. PLoS One 2011; 6:e18690. [PMID: 21533244 PMCID: PMC3076427 DOI: 10.1371/journal.pone.0018690] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2010] [Accepted: 03/15/2011] [Indexed: 11/19/2022] Open
Abstract
Background Human macrophages (Mφ) express low levels of CD4 glycoprotein, which is
constitutively recycled, and 40–50% of its localization is
intracellular at steady-state. Although CD4-interacting proteins in lymphoid
cells are well characterised, little is known about the CD4 protein
interaction-network in human Mφ, which notably lack LCK, a Src family
protein tyrosine kinase believed to stabilise CD4 at the surface of T cells.
As CD4 is the main cellular receptor used by HIV-1, knowledge of its
molecular interactions is important for the understanding of viral infection
strategies. Methodology/Principal Findings We performed large-scale anti-CD4 immunoprecipitations in human primary
Mφ followed by high-resolution mass spectrometry analysis to elucidate
the protein interaction-network involved in induced CD4 internalization and
degradation. Proteomic analysis of CD4 co-immunoisolates in resting Mφ
showed CD4 association with a range of proteins found in the cellular
cortex, membrane rafts and components of clathrin-adaptor proteins, whereas
in induced internalization and degradation CD4 is associated with components
of specific signal transduction, transport and the proteasome. Conclusions/Significance This is the first time that the anti-CD4 co-immunoprecipitation sub-proteome
has been analysed in human primary Mφ. Our data have identified
important Mφ cell surface CD4-interacting proteins, as well as
regulatory proteins involved in internalization and degradation. The data
give valuable insights into the molecular pathways involved in the
regulation of CD4 expression in Mφ and provide candidates/targets for
further biochemical studies.
Collapse
|
34
|
Momboisse F, Houy S, Ory S, Calco V, Bader MF, Gasman S. How important are Rho GTPases in neurosecretion? J Neurochem 2011; 117:623-31. [PMID: 21392006 DOI: 10.1111/j.1471-4159.2011.07241.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Rho GTPases are small GTP binding proteins belonging to the Ras superfamily which act as molecular switches that regulate many cellular function including cell morphology, cell to cell interaction, cell migration and adhesion. In neuronal cells, Rho GTPases have been proposed to regulate neuronal development and synaptic plasticity. However, the role of Rho GTPases in neurosecretion is poorly documented. In this review, we discuss data that highlight the importance of Rho GTPases and their regulators into the control of neurotransmitter and hormone release in neurons and neuroendocrine cells, respectively.
Collapse
Affiliation(s)
- Fanny Momboisse
- CNRS UPR 3212, Institut des Neurosciences Cellulaires et Intégratives, Université de Strasbourg, Strasbourg, France
| | | | | | | | | | | |
Collapse
|
35
|
Schollenberger L, Gronemeyer T, Huber CM, Lay D, Wiese S, Meyer HE, Warscheid B, Saffrich R, Peränen J, Gorgas K, Just WW. RhoA regulates peroxisome association to microtubules and the actin cytoskeleton. PLoS One 2010; 5:e13886. [PMID: 21079737 PMCID: PMC2975642 DOI: 10.1371/journal.pone.0013886] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2010] [Accepted: 10/18/2010] [Indexed: 11/24/2022] Open
Abstract
The current view of peroxisome inheritance provides for the formation of new peroxisomes by both budding from the endoplasmic reticulum and autonomous division. Here we investigate peroxisome-cytoskeleton interactions and show by proteomics, biochemical and immunofluorescence analyses that actin, non-muscle myosin IIA (NMM IIA), RhoA, Rho kinase II (ROCKII) and Rab8 associate with peroxisomes. Our data provide evidence that (i) RhoA in its inactive state, maintained for example by C. botulinum toxin exoenzyme C3, dissociates from peroxisomes enabling microtubule-based peroxisomal movements and (ii) dominant-active RhoA targets to peroxisomes, uncouples the organelles from microtubules and favors Rho kinase recruitment to peroxisomes. We suggest that ROCKII activates NMM IIA mediating local peroxisomal constrictions. Although our understanding of peroxisome-cytoskeleton interactions is still incomplete, a picture is emerging demonstrating alternate RhoA-dependent association of peroxisomes to the microtubular and actin cytoskeleton. Whereas association of peroxisomes to microtubules clearly serves bidirectional, long-range saltatory movements, peroxisome-acto-myosin interactions may support biogenetic functions balancing peroxisome size, shape, number, and clustering.
Collapse
Affiliation(s)
- Lukas Schollenberger
- Heidelberg Center of Biochemistry, University of Heidelberg, Heidelberg, Germany
| | - Thomas Gronemeyer
- Medical Proteom-Center, University of Bochum, Bochum, Germany
- Department for Molecular Genetics and Cell Biology, University of Ulm, Ulm, Germany
| | - Christoph M. Huber
- Heidelberg Center of Biochemistry, University of Heidelberg, Heidelberg, Germany
| | - Dorothee Lay
- Heidelberg Center of Biochemistry, University of Heidelberg, Heidelberg, Germany
| | - Sebastian Wiese
- Medical Proteom-Center, University of Bochum, Bochum, Germany
| | - Helmut E. Meyer
- Medical Proteom-Center, University of Bochum, Bochum, Germany
| | | | - Rainer Saffrich
- Department of Internal Medicine V, University of Heidelberg, Heidelberg, Germany
| | - Johan Peränen
- Institute of Biotechnology, University of Helsinki, Finland
| | - Karin Gorgas
- Department of Anatomy and Medical Cell Biology, University of Heidelberg, Heidelberg, Germany
| | - Wilhelm W. Just
- Heidelberg Center of Biochemistry, University of Heidelberg, Heidelberg, Germany
- * E-mail:
| |
Collapse
|
36
|
Shi L, Fu AK, Ip NY. Multiple roles of the Rho GEF ephexin1 in synapse remodeling. Commun Integr Biol 2010; 3:622-4. [PMID: 21331259 DOI: 10.4161/cib.3.6.13481] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2010] [Accepted: 08/31/2010] [Indexed: 11/19/2022] Open
Abstract
Synapse remodeling, which involves changes in the synaptic structure and their molecular composition, is required for the maturation and refinement of neural circuits. Although synapse remodeling is known to be tightly dependent on the assembly of local actin cytoskeleton, how actin directs the structural changes of synapse and targeting of synaptic proteins are not fully understood. Recently, we identified ephexin1, a Rho guanine nucleotide exchange factor (GEF) that regulates actin dynamics, to play an essential role in the maturation and functioning of the mammalian neuromuscular junction (NMJ). We showed that ephexin1 regulates the synaptic organization of the neurotransmitter receptor acetylcholine receptor (AChR) clusters through RhoA-dependent actin reorganization. Interestingly, ephexin1 has been implicated in the regulation of postsynaptic structure as well as the presynaptic vesicle release at various types of synapses. Our findings thus establish a novel function of ephexin1 in synapse remodeling through regulating the synaptic targeting of neurotransmitter receptors, revealing a versatile role of ephexin1 at synapses.
Collapse
Affiliation(s)
- Lei Shi
- Department of Biochemistry; Molecular Neuroscience Center; State Key Laboratory of Molecular Neuroscience; The Hong Kong University of Science and Technology; Clear Water Bay, Hong Kong, China
| | | | | |
Collapse
|
37
|
McIntosh J, Dennison G, Holly JMP, Jarrett C, Frankow A, Foulstone EJ, Winters ZE, Perks CM. IGFBP-3 can either inhibit or enhance EGF-mediated growth of breast epithelial cells dependent upon the presence of fibronectin. J Biol Chem 2010; 285:38788-800. [PMID: 20851879 DOI: 10.1074/jbc.m110.177311] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Progression of breast cancer is associated with remodeling of the extracellular matrix, often involving a switch from estrogen dependence to a dependence on EGF receptor (EGFR)/HER-2 and is accompanied by increased expression of the main binding protein for insulin-like growth factors (IGFBP-3). We have examined the effects of IGFBP-3 on EGF responses of breast epithelial cells in the context of changes in the extracellular matrix. On plastic and laminin with MCF-10A normal breast epithelial cells, EGF and IGFBP-3 each increased cell growth and together produced a synergistic response, whereas with T47D breast cancer cells IGFBP-3 alone had no effect, but the ability of EGF to increase cell proliferation was markedly inhibited in the presence of IGFBP-3. In contrast on fibronectin with MCF-10A cells, IGFBP-3 alone inhibited cell growth and blocked EGF-induced proliferation. With the cancer cells, IGFBP-3 alone had no effect but enhanced the EGF-induced increase in cell growth. The insulin-like growth factor-independent effects of IGFBP-3 alone on cell proliferation were completely abrogated in the presence of an EGFR, tyrosine kinase inhibitor, Iressa. Although IGFBP-3 did not affect EGFR phosphorylation [Tyr(1068)], it was found to modulate receptor internalization and was associated with activation of Rho and subsequent changes in MAPK phosphorylation. The levels of fibronectin and IGFBP-3 within breast tumors may determine their dependence on EGFR and their response to therapies targeting this receptor.
Collapse
Affiliation(s)
- Jamie McIntosh
- School of Clinical Sciences, IGFs and Metabolic Endocrinology Group, Learning and Research Building, 2nd Floor, University of Bristol, Southmead Hospital, Bristol BS10 5NB, United Kingdom
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Bu W, Lim KB, Yu YH, Chou AM, Sudhaharan T, Ahmed S. Cdc42 interaction with N-WASP and Toca-1 regulates membrane tubulation, vesicle formation and vesicle motility: implications for endocytosis. PLoS One 2010; 5:e12153. [PMID: 20730103 PMCID: PMC2921345 DOI: 10.1371/journal.pone.0012153] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2010] [Accepted: 07/12/2010] [Indexed: 11/18/2022] Open
Abstract
Transducer of Cdc42-dependent actin assembly (Toca-1) consists of an F-BAR domain, a Cdc42 binding site and an SH3 domain. Toca-1 interacts with N-WASP, an activator of actin nucleation that binds Cdc42. Cdc42 may play an important role in regulating Toca-1 and N-WASP functions. We report here that the cellular expression of Toca-1 and N-WASP induces membrane tubulation and the formation of motile vesicles. Marker and uptake analysis suggests that the tubules and vesicles are associated with clathrin-mediated endocytosis. Forster resonance energy transfer (FRET) and Fluorescence Lifetime Imaging Microscopy (FLIM) analysis shows that Cdc42, N-WASP and Toca-1 form a trimer complex on the membrane tubules and vesicles and that Cdc42 interaction with N-WASP is critical for complex formation. Modulation of Cdc42 interaction with Toca-1 and/or N-WASP affects membrane tubulation, vesicle formation and vesicle motility. Thus Cdc42 may influence endocytic membrane trafficking by regulating the formation and activity of the Toca-1/N-WASP complex.
Collapse
Affiliation(s)
- Wenyu Bu
- Neural Stem Cell Laboratory, Institute of Medical Biology, Singapore, Singapore
| | - Kim Buay Lim
- Neural Stem Cell Laboratory, Institute of Medical Biology, Singapore, Singapore
| | - Yuan Hong Yu
- Neural Stem Cell Laboratory, Institute of Medical Biology, Singapore, Singapore
| | - Ai Mei Chou
- Neural Stem Cell Laboratory, Institute of Medical Biology, Singapore, Singapore
| | - Thankiah Sudhaharan
- Neural Stem Cell Laboratory, Institute of Medical Biology, Singapore, Singapore
| | - Sohail Ahmed
- Neural Stem Cell Laboratory, Institute of Medical Biology, Singapore, Singapore
- * E-mail:
| |
Collapse
|
39
|
Zonia L. Spatial and temporal integration of signalling networks regulating pollen tube growth. JOURNAL OF EXPERIMENTAL BOTANY 2010; 61:1939-57. [PMID: 20378665 DOI: 10.1093/jxb/erq073] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Abstract
The overall function of a cell is determined by its contingent of active signal transduction cascades interacting on multiple levels with metabolic pathways, cytoskeletal organization, and regulation of gene expression. Much work has been devoted to analysis of individual signalling cascades interacting with unique cellular targets. However, little is known about how cells integrate information across hierarchical signalling networks. Recent work on pollen tube growth indicates that several key signalling cascades respond to changes in cell hydrodynamics and apical volume. Combined with known effects on cytoarchitecture and signalling from other cell systems, hydrodynamics has the potential to integrate and synchronize the function of the broader signalling network in pollen tubes. This review will explore recent work on cell hydrodynamics in a variety of systems including pollen, and discuss hydrodynamic regulation of cell signalling and function including exocytosis and endocytosis, actin cytoskeleton reorganization, cell wall deposition and assembly, phospholipid and inositol polyphosphate signalling, ion flux, small G-proteins, fertilization, and self-incompatibility. The combined data support a newly emerging model of pollen tube growth.
Collapse
Affiliation(s)
- Laura Zonia
- University of Amsterdam, Swammerdam Institute for Life Sciences, Section of Plant Physiology, Kruislaan 904, 1098 XH Amsterdam, The Netherlands.
| |
Collapse
|
40
|
Samson T, Welch C, Monaghan-Benson E, Hahn KM, Burridge K. Endogenous RhoG is rapidly activated after epidermal growth factor stimulation through multiple guanine-nucleotide exchange factors. Mol Biol Cell 2010; 21:1629-42. [PMID: 20237158 PMCID: PMC2861620 DOI: 10.1091/mbc.e09-09-0809] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
In this article it is shown that EGF stimulation leads to rapid activation of RhoG through Vav GEFs and the GEF PLEKHG6. Importantly, different cellular responses induced by EGF are determined by the available GEFs. Furthermore, this article presents results showing that EGF-stimulated cell migration and EGFR internalization are regulated by RhoG. RhoG is a member of the Rac-like subgroup of Rho GTPases and has been linked to a variety of different cellular functions. Nevertheless, many aspects of RhoG upstream and downstream signaling remain unclear; in particular, few extracellular stimuli that modulate RhoG activity have been identified. Here, we describe that stimulation of epithelial cells with epidermal growth factor leads to strong and rapid activation of RhoG. Importantly, this rapid activation was not observed with other growth factors tested. The kinetics of RhoG activation after epidermal growth factor (EGF) stimulation parallel the previously described Rac1 activation. However, we show that both GTPases are activated independently of one another. Kinase inhibition studies indicate that the rapid activation of RhoG and Rac1 after EGF treatment requires the activity of the EGF receptor kinase, but neither phosphatidylinositol 3-kinase nor Src kinases. By using nucleotide-free RhoG pull-down assays and small interfering RNA-mediated knockdown studies, we further show that guanine-nucleotide exchange factors (GEFs) of the Vav family mediate EGF-induced rapid activation of RhoG. In addition, we found that in certain cell types the recently described RhoG GEF PLEKHG6 can also contribute to the rapid activation of RhoG after EGF stimulation. Finally, we present results that show that RhoG has functions in EGF-stimulated cell migration and in regulating EGF receptor internalization.
Collapse
Affiliation(s)
- Thomas Samson
- Department of Cell and Developmental Biology, Lineberger Comprehensive Cancer Center, Chapel Hill, NC 27599, USA
| | | | | | | | | |
Collapse
|
41
|
Shi L, Butt B, Ip FCF, Dai Y, Jiang L, Yung WH, Greenberg ME, Fu AKY, Ip NY. Ephexin1 is required for structural maturation and neurotransmission at the neuromuscular junction. Neuron 2010; 65:204-16. [PMID: 20152127 DOI: 10.1016/j.neuron.2010.01.012] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/24/2009] [Indexed: 12/24/2022]
Abstract
The maturation of neuromuscular junctions (NMJs) requires the topological transformation of postsynaptic acetylcholine receptor (AChR)-containing structures from a simple plaque to an elaborate structure composed of pretzel-like branches. This maturation process results in the precise apposition of the presynaptic and postsynaptic specializations. However, little is known about the molecular mechanisms underlying the plaque-to-pretzel transition of AChR clusters. In this study, we identify an essential role for the RhoGEF ephexin1 in the maturation of AChR clusters. Adult ephexin1(-/-) mice exhibit severe muscle weakness and impaired synaptic transmission at the NMJ. Intriguingly, when ephexin1 expression is deficient in vivo, the NMJ fails to mature into the pretzel-like shape, and such abnormalities can be rescued by re-expression of ephexin1. We further demonstrate that ephexin1 regulates the stability of AChR clusters in a RhoA-dependent manner. Taken together, our findings reveal an indispensible role for ephexin1 in regulating the structural maturation and neurotransmission of NMJs.
Collapse
Affiliation(s)
- Lei Shi
- Department of Biochemistry, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong SAR, China
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Manfredini F, Benati D, Beani L. The strepsipteran endoparasite Xenos vesparum alters the immunocompetence of its host, the paper wasp Polistes dominulus. JOURNAL OF INSECT PHYSIOLOGY 2010; 56:253-259. [PMID: 19879278 DOI: 10.1016/j.jinsphys.2009.10.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/22/2009] [Revised: 10/19/2009] [Accepted: 10/21/2009] [Indexed: 05/28/2023]
Abstract
It is unexplained how strepsipteran insects manipulate the physiology of their hosts in order to undergo endoparasitic development without being entrapped by the innate immune defences of the host. Here we present pioneering work that aimed to explore for the first time several components of the cellular and humoral immune response among immature stages of the paper wasp Polistes dominulus, in both unparasitized insects and after infection by the strepsipteran endoparasite Xenos vesparum. We carried out hemocyte counts, phagocytosis assays in vitro and antibacterial response in vivo. On the whole, hemocyte load does not seem to be drastically affected by parasitization: a non-significant increase in hemocyte numbers was observed in parasitized wasps as respect to control, while the two dominant hemocyte types were present with similar proportions in both groups. On the other hand, phagocytosis was significantly reduced in hemocytes from parasitized wasps while the antibacterial response seemed to be less effective in control. These somewhat unexpected results are discussed, along with the implications of a multiple approach in immune response studies.
Collapse
Affiliation(s)
- Fabio Manfredini
- Department of Evolutionary Biology, University of Siena, via Aldo Moro 2, 53100 Siena, Italy.
| | | | | |
Collapse
|
43
|
Guanine Exchange Factor Vav2: A Novel Potential Target for the Development of Drugs Effective in the Prevention of Papillomavirus Infection and Disease. Am J Ther 2009; 16:496-507. [DOI: 10.1097/mjt.0b013e31819be0a5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
44
|
Koike S, Keino-Masu K, Ohto T, Sugiyama F, Takahashi S, Masu M. Autotaxin/lysophospholipase D-mediated lysophosphatidic acid signaling is required to form distinctive large lysosomes in the visceral endoderm cells of the mouse yolk sac. J Biol Chem 2009; 284:33561-70. [PMID: 19808661 DOI: 10.1074/jbc.m109.012716] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Autotaxin, a lysophospholipase D encoded by the Enpp2 gene, is an exoenzyme that produces lysophosphatidic acid in the extracellular space. Lysophosphatidic acid acts on specific G protein-coupled receptors, thereby regulating cell growth, migration, and survival. Previous studies have revealed that Enpp2(-/-) mouse embryos die at about embryonic day (E) 9.5 because of angiogenic defects in the yolk sac. However, what cellular defects occur in Enpp2(-/-) embryos and what intracellular signaling pathways are involved in the phenotype manifestation remain unknown. Here, we show that Enpp2 is required to form distinctive large lysosomes in the yolk sac visceral endoderm cells. From E7.5 to E9.5, Enpp2 mRNA is abundantly expressed in the visceral endoderm cells. In Enpp2(-/-) mouse embryos, lysosomes in the visceral endoderm cells are fragmented. By using a whole embryo culture system combined with specific pharmacological inhibitors for intracellular signaling molecules, we show that lysophosphatidic acid receptors and the Rho-Rho-associated coiled-coil containing protein kinase (ROCK)-LIM kinase pathway are required to form large lysosomes. In addition, electroporation of dominant negative forms of Rho, ROCK, or LIM kinase also leads to the size reduction of lysosomes in wild-type visceral endoderm cells. In Enpp2(-/-) visceral endoderm cells, the steady-state levels of cofilin phosphorylation and actin polymerization are reduced. In addition, perturbations of actin turnover dynamics by actin inhibitors cytochalasin B and jasplakinolide result in the defect in lysosome formation. These results suggest that constitutive activation of the Rho-ROCK-LIM kinase pathway by extracellular production of lysophosphatidic acid by the action of autotaxin is required to maintain the large size of lysosomes in visceral endoderm cells.
Collapse
Affiliation(s)
- Seiichi Koike
- Department of Molecular Neurobiology, Institute of Basic Medical Sciences, University of Tsukuba, 1-1-1 Tennoudai, Tsukuba, Ibaraki 305-8577, Japan
| | | | | | | | | | | |
Collapse
|
45
|
Actin dynamics and Rho GTPases regulate the size and formation of parasitophorous vacuoles containing Coxiella burnetii. Infect Immun 2009; 77:4609-20. [PMID: 19635823 DOI: 10.1128/iai.00301-09] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Q fever is a disease caused by Coxiella burnetii. In the host cell, this pathogen generates a large parasitophorous vacuole (PV) with lysosomal characteristics. Here we show that F-actin not only is recruited to but also is involved in the formation of the typical PV. Treatment of infected cells with F-actin-depolymerizing agents alters PV development. The small PVs formed in latrunculin B-treated cells were loaded with transferrin and Lysotracker and labeled with an antibody against cathepsin D, suggesting that latrunculin B did not affect vacuole cargo and its lysosomal characteristics. Nevertheless, the vacuoles were unable to fuse with latex bead phagosomes. It is known that actin dynamics are regulated by the Rho family GTPases. To assess the role of these GTPases in PV formation, infected cells were transfected with pEGFP expressing wild-type and mutant Rac1, Cdc42, and RhoA proteins. Rac1 did not show significant PV association. In contrast, PVs were decorated by both the wild types and constitutively active mutants of Cdc42 and RhoA. This association was inhibited by treatment of infected cells with chloramphenicol, suggesting a role for bacterial protein synthesis in the recruitment of these proteins. Interestingly, a decrease in vacuole size was observed in cells expressing dominant-negative RhoA; however, these small vacuoles accumulated transferrin, Lysotracker, and DQ-BSA. In summary, these results suggest that actin, likely modulated by the GTPases RhoA and Cdc42 and by bacterial proteins, is involved in the formation of the typical PV.
Collapse
|
46
|
Yacobi NR, Malmstadt N, Fazlollahi F, DeMaio L, Marchelletta R, Hamm-Alvarez SF, Borok Z, Kim KJ, Crandall ED. Mechanisms of alveolar epithelial translocation of a defined population of nanoparticles. Am J Respir Cell Mol Biol 2009; 42:604-14. [PMID: 19574531 DOI: 10.1165/rcmb.2009-0138oc] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
To explore mechanisms of nanoparticle interactions with and trafficking across lung alveolar epithelium, we utilized primary rat alveolar epithelial cell monolayers (RAECMs) and an artificial lipid bilayer on filter model (ALBF). Trafficking rates of fluorescently labeled polystyrene nanoparticles (PNPs; 20 and 100 nm, carboxylate (negatively charged) or amidine (positively charged)-modified) in the apical-to-basolateral direction under various experimental conditions were measured. Using confocal laser scanning microscopy, we investigated PNP colocalization with early endosome antigen-1, caveolin-1, clathrin heavy chain, cholera toxin B, and wheat germ agglutinin. Leakage of 5-carboxyfluorescein diacetate from RAECMs, and trafficking of (22)Na and (14)C-mannitol across ALBF, were measured in the presence and absence of PNPs. Results showed that trafficking of positively charged PNPs was 20-40 times that of negatively charged PNPs across both RAECMs and ALBF, whereas translocation of PNPs across RAECMs was 2-3 times faster than that across ALBF. Trafficking rates of PNPs across RAECMs did not change in the presence of EGTA (which decreased transepithelial electrical resistance to zero) or inhibitors of endocytosis. Confocal laser scanning microscopy revealed no intracellular colocalization of PNPs with early endosome antigen-1, caveolin-1, clathrin heavy chain, cholera toxin B, or wheat germ agglutinin. Leakage of 5-carboxyfluorescein diacetate from alveolar epithelial cells, and sodium ion and mannitol flux across ALBF, were not different in the presence or absence of PNPs. These data indicate that PNPs translocate primarily transcellularly across RAECMs, but not via known major endocytic pathways, and suggest that such translocation may take place by diffusion of PNPs through the lipid bilayer of cell plasma membranes.
Collapse
Affiliation(s)
- Nazanin R Yacobi
- Mork Family Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, California 90033, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Lee S, Han JW, Leeper L, Gruver JS, Chung CY. Regulation of the formation and trafficking of vesicles from Golgi by PCH family proteins during chemotaxis. BIOCHIMICA ET BIOPHYSICA ACTA 2009; 1793:1199-209. [PMID: 19409937 PMCID: PMC2703453 DOI: 10.1016/j.bbamcr.2009.04.012] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 11/04/2008] [Revised: 04/22/2009] [Accepted: 04/24/2009] [Indexed: 12/14/2022]
Abstract
Previous study demonstrated that WASP localizes on vesicles during Dictyostelium chemotaxis and these vesicles appear to be preferentially distributed at the leading and trailing edge of migrating cells. In this study, we have examined the role of PCH family proteins, Nwk/Bzz1p-like protein (NLP) and Syndapin-like protein (SLP), in the regulation of the formation and trafficking of WASP-vesicles during chemotaxis. NLP and SLP appear to be functionally redundant and deletion of both nlp and slp genes causes the loss of polarized F-actin organization and significant defects in chemotaxis. WASP and NLP are colocalized on vesicles and interactions between two molecules via the SH3 domain of NLP/SLP and the proline-rich repeats of WASP are required for vesicle formation from Golgi. Microtubules are required for polarized trafficking of these vesicles as vesicles showing high directed mobility are absent in cells treated with nocodazole. Our results suggest that interaction of WASP with NLP/SLP is required for the formation and trafficking of vesicles from Golgi to the membrane, which might play a central role in the establishment of cell polarity during chemotaxis.
Collapse
Affiliation(s)
- S. Lee
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37232-6600
| | - J. W. Han
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37232-6600
| | - L. Leeper
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37232-6600
| | - J. S. Gruver
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37232-6600
| | - C. Y. Chung
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37232-6600
| |
Collapse
|
48
|
Zonia L, Munnik T. Uncovering hidden treasures in pollen tube growth mechanics. TRENDS IN PLANT SCIENCE 2009; 14:318-27. [PMID: 19446491 DOI: 10.1016/j.tplants.2009.03.008] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/03/2008] [Revised: 02/22/2009] [Accepted: 03/03/2009] [Indexed: 05/08/2023]
Abstract
The long-standing model of tip growth in pollen tubes considers that exocytosis and growth occur at the apex and that the pool of very small vesicles in the apical dome contains secretory (exocytic) vesicles. However, recent work on vesicle trafficking dynamics in tobacco pollen tubes shows that exocytosis occurs in the subapical region. Taking these and other new results into account, we set out to resolve specific problems that are endemic in current models and present a two-part ACE (apical cap extension)-H (hydrodynamics) growth model. The ACE model involves delivery and recycling of materials required for new cell synthesis and the H model involves mechanisms that integrate and regulate key cellular pathways and drive cell elongation during growth.
Collapse
Affiliation(s)
- Laura Zonia
- Swammerdam Institute for Life Sciences, Plant Physiology Section, University of Amsterdam, Kruislaan 904, 1098 XH Amsterdam, The Netherlands.
| | | |
Collapse
|
49
|
Bu W, Chou AM, Lim KB, Sudhaharan T, Ahmed S. The Toca-1-N-WASP complex links filopodial formation to endocytosis. J Biol Chem 2009; 284:11622-36. [PMID: 19213734 DOI: 10.1074/jbc.m805940200] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The transducer of Cdc42-dependent actin assembly (Toca-1)-N-WASP complex was isolated as an essential cofactor for Cdc42-driven actin polymerization in vitro. Toca-1 consists of an N-terminal F-BAR domain, followed by a Cdc42 binding site (HR1 domain) and an SH3 domain, (the N-WASP interacting site). N-WASP is an activator of actin nucleation through the Arp2/3 complex. The aim of the present study was to investigate the cellular function of the Toca-1-N-WASP complex. We report that Toca-1 induces filopodia and neurites as does N-WASP in N1E115 neuroblastoma cells. Toca-1 requires the F-BAR domain, Cdc42 binding site, and SH3 domain to induce filopodia. Toca-1 and N-WASP both require each other to induce filopodia. The expression of Toca-1 and N-WASP affects the distribution, size, and number of Rab5 positive membranes. Toca-1 interacts directly with N-WASP in filopodia and Rab5 membrane as seen by Forster resonance energy transfer. Thus the Toca-1-N-WASP complex localizes to and induces the formation of filopodia and endocytic vesicles. Last, three inhibitors of endocytosis, Dynamin-K44A, Eps15Delta95/295, and clathrin heavy chain RNA interference, block Toca-1-induced filopodial formation. Taken together, these data suggest that the Toca-1-N-WASP complex can link filopodial formation to endocytosis.
Collapse
Affiliation(s)
- Wenyu Bu
- Institute of Medical Biology, 8A Biomedical Grove, Immunos, Singapore 138665
| | | | | | | | | |
Collapse
|
50
|
Pridgeon JW, Webber EA, Sha D, Li L, Chin LS. Proteomic analysis reveals Hrs ubiquitin-interacting motif-mediated ubiquitin signaling in multiple cellular processes. FEBS J 2009; 276:118-31. [PMID: 19019082 PMCID: PMC2647816 DOI: 10.1111/j.1742-4658.2008.06760.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Despite the critical importance of protein ubiquitination in the regulation of diverse cellular processes, the molecular mechanisms by which cells recognize and transmit ubiquitin signals remain poorly understood. The endosomal sorting machinery component hepatocyte growth factor-regulated tyrosine kinase substrate (Hrs) contains a ubiquitin-interacting motif (UIM), which is believed to bind ubiquitinated membrane cargo proteins and mediate their sorting to the lysosomal degradation pathway. To gain insight into the role of Hrs UIM-mediated ubiquitin signaling in cells, we performed a proteomic screen for Hrs UIM-interacting ubiquitinated proteins in human brain by using an in vitro expression cloning screening approach. We have identified 48 ubiquitinated proteins that are specifically recognized by the UIM domain of Hrs. Among them, 12 are membrane proteins that are likely to be Hrs cargo proteins, and four are membrane protein-associated adaptor proteins whose ubiquitination may act as a signal to target their associated membrane cargo for Hrs-mediated endosomal sorting. Other classes of the identified proteins include components of the vesicular trafficking machinery, cell signaling molecules, proteins associated with the cytoskeleton and cytoskeleton-dependent transport, and enzymes involved in ubiquitination and metabolism, suggesting the involvement of Hrs UIM-mediated ubiquitin signaling in the regulation of multiple cellular processes. We have characterized the ubiquitination of two identified proteins, Munc18-1 and Hsc70, and their interaction with Hrs UIM, and provided functional evidence supporting a role for Hsc70 in the regulation of Hrs-mediated endosome-to-lysosome trafficking.
Collapse
Affiliation(s)
- Julia W Pridgeon
- Department of Pharmacology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | | | | | | | | |
Collapse
|