1
|
Cao Q, Zhang W, Liu X, Li Y. AtFTCD-L, a trans-Golgi network localized protein, modulates root growth of Arabidopsis in high-concentration agar culture medium. PLANTA 2022; 256:3. [PMID: 35637390 DOI: 10.1007/s00425-022-03911-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 05/07/2022] [Indexed: 06/15/2023]
Abstract
AtFTCD-L protein is localized on the TGN vesicles in Arabidopsis root cap cells. AtFTCD-L mutation resulted in slow root growth of Arabidopsis in high-concentration agar culture medium. Arabidopsis formiminotransferase cyclodeaminase-like protein (AtFTCD-L) in Arabidopsis is homologous to the formiminotransferase cyclodeaminase (FTCD) protein in animal cells. However, the localization and function of AtFTCD-L remain unknown in Arabidopsis. In this study, we generated and analyzed a deletion mutant of AtFTCD-L with a T-DNA insertion. We found that the growth of Arabidopsis roots with the T-DNA insertion mutation in AtFTCD-L was slower than that of wild-type roots when grown in high-concentration 1/2 MS agar culture medium. AtFTCD-L-GFP could restore the ftcd-l mutant phenotype. In addition, the AtFTCD-L protein was localized on the trans-Golgi network (TGN) vesicles in Arabidopsis root cap cells. Fluorescence recovery after photobleaching (FRAP) experiment using Arabidopsis pollen-specific receptor-like kinase-GFP (AtPRK1-GFP) stably transformed plants showed that the deficiency of AtFTCD-L protein in Arabidopsis led to slower secretion in the root cap peripheral cells. The AtFTCD-L protein deficiency also resulted in a significantly reduced monosaccharides content in the culture medium. Based on the above results, we speculate that the AtFTCD-L protein may be involved in sorting and/or transportation of TGN vesicles in root cap peripheral cells, thereby regulating the extracellular secretion of mucilage components in the root cap.
Collapse
Affiliation(s)
- Qijiang Cao
- State Key Laboratory of Plant Physiology and Biochemistry, College of Biological Sciences, China Agricultural University, Beijing, 100193, China
- College of Life Sciences and Engineering, Shenyang University, Liaoning, 110044, China
| | - Wei Zhang
- State Key Laboratory of Plant Physiology and Biochemistry, College of Biological Sciences, China Agricultural University, Beijing, 100193, China
| | - Xinyan Liu
- State Key Laboratory of Plant Physiology and Biochemistry, College of Biological Sciences, China Agricultural University, Beijing, 100193, China
| | - Yan Li
- State Key Laboratory of Plant Physiology and Biochemistry, College of Biological Sciences, China Agricultural University, Beijing, 100193, China.
| |
Collapse
|
2
|
Wu Y, Bai Y, McEwan DG, Bentley L, Aravani D, Cox RD. Palmitoylated small GTPase ARL15 is translocated within Golgi network during adipogenesis. Biol Open 2021; 10:273707. [PMID: 34779483 PMCID: PMC8689486 DOI: 10.1242/bio.058420] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 11/10/2021] [Indexed: 11/20/2022] Open
Abstract
The small GTPase ARF family member ARL15 gene locus is associated in population studies with increased risk of type 2 diabetes, lower adiponectin and higher fasting insulin levels. Previously, loss of ARL15 was shown to reduce insulin secretion in a human β-cell line and loss-of-function mutations are found in some lipodystrophy patients. We set out to understand the role of ARL15 in adipogenesis and showed that endogenous ARL15 palmitoylated and localised in the Golgi of mouse liver. Adipocyte overexpression of palmitoylation-deficient ARL15 resulted in redistribution to the cytoplasm and a mild reduction in expression of some adipogenesis-related genes. Further investigation of the localisation of ARL15 during differentiation of a human white adipocyte cell line showed that ARL15 was predominantly co-localised with a marker of the cis face of Golgi at the preadipocyte stage and then translocated to other Golgi compartments after differentiation was induced. Finally, co-immunoprecipitation and mass spectrometry identified potential interacting partners of ARL15, including the ER-localised protein ARL6IP5. Together, these results suggest a palmitoylation dependent trafficking-related role of ARL15 as a regulator of adipocyte differentiation via ARL6IP5 interaction. This article has an associated First Person interview with the first author of the paper. Summary: ARL15 (GTPase ARF family) is associated with adipose traits. ARL15 is palmitoylated, localised to Golgi in preadipocytes and translocated to other Golgi compartments during differentiation. ARL15 interacts with ER-localised ARL6IP5.
Collapse
Affiliation(s)
- Yixing Wu
- Mammalian Genetics Unit, MRC Harwell Institute, Harwell Oxford, Oxfordshire, OX11 0RD, UK
| | - Ying Bai
- Mammalian Genetics Unit, MRC Harwell Institute, Harwell Oxford, Oxfordshire, OX11 0RD, UK
| | - David G McEwan
- Division of Cell Signalling & Immunology, School of Life Sciences, University of Dundee, Dundee, UK.,Cancer Research UK Beatson Institute, Garscube Estate, Switchback Road, Glasgow, G61 1BD, UK
| | - Liz Bentley
- Mammalian Genetics Unit, MRC Harwell Institute, Harwell Oxford, Oxfordshire, OX11 0RD, UK
| | - Dimitra Aravani
- Mammalian Genetics Unit, MRC Harwell Institute, Harwell Oxford, Oxfordshire, OX11 0RD, UK
| | - Roger D Cox
- Mammalian Genetics Unit, MRC Harwell Institute, Harwell Oxford, Oxfordshire, OX11 0RD, UK
| |
Collapse
|
3
|
Identification of Hub Genes and Analysis of Prognostic Values in Hepatocellular Carcinoma by Bioinformatics Analysis. Am J Med Sci 2020; 359:226-234. [PMID: 32200915 DOI: 10.1016/j.amjms.2020.01.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 12/25/2019] [Accepted: 01/14/2020] [Indexed: 12/16/2022]
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is one of the most frequent cancers in the world. In this study, differentially expressed genes (DEGs) between tumor tissues and normal tissues were identified using the comprehensive analysis method in bioinformatics. MATERIALS AND METHODS We downloaded 3 mRNA expression profiles from the Gene Expression Omnibus database to identify DEGs between tumor tissues and adjacent normal tissues. The Gene Ontology, Kyoto Encyclopedia of Genes and Genomes pathway analysis, protein-protein interaction network was performed to understand the function of DEGs. OncoLnc, which was linked to The Cancer Genome Atlas survival data, was used to investigate the prognostic values of hub genes. The expression of selected hub genes was validated by the quantitative real-time polymerase chain reaction. RESULTS A total of 235 DEGs, consisting of 36 upregulated and 199 downregulated genes, were identified between tumor tissue and normal tissue. The Gene Ontology and Kyoto Encyclopedia of Genes and Genomes analysis results showed the upregulated DEGs to be significantly enriched in cell division, mid-body, ATP binding and oocyte meiosis pathways. The downregulated DEGs were mainly involved in epoxygenase P450 pathway, extracellular region, oxidoreductase activity and metabolic pathways. Ten hub genes, including Aurora kinase A, Cell division cycle 20, formiminotransferase cyclodeaminase, UBE2C, Cyclin B2, pituitary tumor-transforming gene 1, CDKN3, CKS1B, Topoisomerase-II alpha and KIF20A, were identified as the key genes in HCC. Survival analysis found the expression of hub genes to be significantly correlated with the survival of patients with HCC. CONCLUSIONS The present study identified hub genes and pathways in HCC that may be potential targets for diagnosis, treatment and prognostic prediction.
Collapse
|
4
|
US3 Kinase-Mediated Phosphorylation of Tegument Protein VP8 Plays a Critical Role in the Cellular Localization of VP8 and Its Effect on the Lipid Metabolism of Bovine Herpesvirus 1-Infected Cells. J Virol 2019; 93:JVI.02151-18. [PMID: 30626671 DOI: 10.1128/jvi.02151-18] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Accepted: 12/11/2018] [Indexed: 02/04/2023] Open
Abstract
Bovine herpesvirus 1 (BoHV-1) infects bovine species, causing respiratory infections, genital disorders and abortions. VP8 is the most abundant tegument protein of BoHV-1 and is critical for virus replication in cattle. In this study, the cellular transport of VP8 in BoHV-1-infected cells and its ability to alter the cellular lipid metabolism were investigated. A viral kinase, US3, was found to be involved in regulating these processes. In the early stages of infection VP8 was localized in the nucleus. Subsequently, presumably after completion of its role in the nucleus, VP8 was translocated to the cytoplasm. When US3 was deleted or the essential US3 phosphorylation site of VP8 was mutated in BoHV-1, the majority of VP8 was localized in the nuclei of infected cells. This suggests that phosphorylation by US3 may be critical for cytoplasmic localization of VP8. Eventually, the cytoplasmic VP8 was accumulated in the cis-Golgi apparatus but not in the trans-Golgi network, implying that VP8 was not involved in virion transport toward and budding from the cell membrane. VP8 caused lipid droplet (LD) formation in the nuclei of transfected cells and increased cellular cholesterol levels. Lipid droplets were not found in the nuclei of BoHV-1-infected cells when VP8 was cytoplasmic in the presence of US3. However, when US3 was deleted or phosphorylation residues in VP8 were mutated, nuclear VP8 and LDs appeared in BoHV-1-infected cells. The total cholesterol level was increased in BoHV-1-infected cells but not in ΔUL47-BoHV-1-infected cells, further supporting a role for VP8 in altering the cellular lipid metabolism during infection.IMPORTANCE Nuclear localization signals (NLSs) and nuclear export signals (NESs) are important elements directing VP8 to the desired locations in the BoHV-1-infected cell. In this study, a critical regulator that switches the nuclear and cytoplasmic localization of VP8 in BoHV-1-infected cells was identified. BoHV-1 used viral kinase US3 to regulate the cellular localization of VP8. Early during BoHV-1 infection VP8 was localized in the nucleus, where it performs various functions; once US3 was expressed, phosphorylated VP8 was cytoplasmic and ultimately accumulated in the cis-Golgi apparatus, presumably to be incorporated into virions. The Golgi localization of VP8 was only observed in virus-infected cells and not in US3-cotransfected cells, suggesting that this is mediated by other viral factors. Interestingly, VP8 was shown to cause increased cholesterol levels, which is a novel function for VP8 and a potential strategy to supply lipid for viral replication.
Collapse
|
5
|
Infectious Bursal Disease Virus Hijacks Endosomal Membranes as the Scaffolding Structure for Viral Replication. J Virol 2018. [PMID: 29540593 DOI: 10.1128/jvi.01964-17] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Birnaviruses are unconventional members of the group of double-stranded RNA (dsRNA) viruses that are characterized by the lack of a transcriptionally active inner core. Instead, the birnaviral particles organize their genome in ribonucleoprotein complexes (RNPs) composed by dsRNA segments, the dsRNA-binding VP3 protein, and the virally encoded RNA-dependent RNA polymerase (RdRp). This and other structural features suggest that birnaviruses may follow a completely different replication program from that followed by members of the Reoviridae family, supporting the hypothesis that birnaviruses are the evolutionary link between single-stranded positive RNA (+ssRNA) and dsRNA viruses. Here we demonstrate that infectious bursal disease virus (IBDV), a prototypical member of the Birnaviridae family, hijacks endosomal membranes of infected cells through the interaction of a viral protein, VP3, with the phospholipids on the cytosolic leaflet of these compartments for replication. Employing a mutagenesis approach, we demonstrated that VP3 domain PATCH 2 (P2) mediates the association of VP3 with the endosomal membranes. To determine the role of VP3 P2 in the context of the virus replication cycle, we used avian cells stably overexpressing VP3 P2 for IBDV infection. Importantly, the intra- and extracellular virus yields, as well as the intracellular levels of VP2 viral capsid protein, were significantly diminished in cells stably overexpressing VP3 P2. Together, our results indicate that the association of VP3 with endosomes has a relevant role in the IBDV replication cycle. This report provides direct experimental evidence for membranous compartments such as endosomes being required by a dsRNA virus for its replication. The results also support the previously proposed role of birnaviruses as an evolutionary link between +ssRNA and dsRNA viruses.IMPORTANCE Infectious bursal disease (IBD; also called Gumboro disease) is an acute, highly contagious immunosuppressive disease that affects young chickens and spreads worldwide. The etiological agent of IBD is infectious bursal disease virus (IBDV). This virus destroys the central immune organ (bursa of Fabricius), resulting in immunosuppression and reduced responses of chickens to vaccines, which increase their susceptibility to other pathogens. IBDV is a member of Birnaviridae family, which comprises unconventional members of dsRNA viruses, whose replication strategy has been scarcely studied. In this report we show that IBDV hijacks the endosomes of the infected cells for establishing viral replication complexes via the association of the ribonucleoprotein complex component VP3 with the phospholipids in the cytosolic leaflet of endosomal membranes. We show that this interaction is mediated by the VP3 PATCH 2 domain and demonstrate its relevant role in the context of viral infection.
Collapse
|
6
|
Busby T, Meissner JM, Styers ML, Bhatt J, Kaushik A, Hjelmeland AB, Sztul E. The Arf activator GBF1 localizes to plasma membrane sites involved in cell adhesion and motility. CELLULAR LOGISTICS 2017; 7:e1308900. [PMID: 28702273 DOI: 10.1080/21592799.2017.1308900] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Revised: 02/20/2017] [Accepted: 03/14/2017] [Indexed: 10/24/2022]
Affiliation(s)
- Theodore Busby
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Justyna M Meissner
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Melanie L Styers
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Jay Bhatt
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Akhil Kaushik
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Anita B Hjelmeland
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Elizabeth Sztul
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
7
|
Carvalho-Oliveira I, Efthymiadou A, Malhó R, Nogueira P, Tzetis M, Kanavakis E, Amaral MD, Penque D. CFTR Localization in Native Airway Cells and Cell Lines Expressing Wild-type or F508del-CFTR by a Panel of Different Antibodies. J Histochem Cytochem 2016; 52:193-203. [PMID: 14729871 DOI: 10.1177/002215540405200207] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
The intracellular localization of cystic fibrosis transmembrane conductance regulator (CFTR) in native tissues is a major issue in the study of mutation, processing, and trafficking effects in CFTR and in the evaluation of therapeutic strategies in cystic fibrosis (CF). This work evaluated the applicability of ten different antibodies (Abs) under various fixation techniques for CFTR localization in fresh-brushed nasal epithelial cells collected from CF patients homozygous for F508del and control individuals. In parallel, the same Ab panel was also tested on BHK cell lines overexpressing wild-type or F508del CFTR. The Abs MATG1061, 169, Lis1, MP-CT1, CC24-R, MAB25031, and MAB1660 gave the best detection of CFTR in the apical region (AR) of nasal tall columnar epithelial (TCE) cells. The labeling pattern of these Abs was consistent with the postulated processing defect of F508del CFTR because only a minority of CF TCE cells present CFTR in the AR. In contrast, M3A7, MM13–4, and L12B4 weakly react with the AR and stain almost exclusively a cis-Golgi-like structure in the majority of CF and non-CF airway cells. In BHK cells, all the Abs enabled distinction between wild-type CFTR localization in cell membrane from F508del CFTR, which in these cells is exclusively located in the endoplasmic reticulum.
Collapse
|
8
|
Guichet PO, Guelfi S, Ripoll C, Teigell M, Sabourin JC, Bauchet L, Rigau V, Rothhut B, Hugnot JP. Asymmetric Distribution of GFAP in Glioma Multipotent Cells. PLoS One 2016; 11:e0151274. [PMID: 26953813 PMCID: PMC4783030 DOI: 10.1371/journal.pone.0151274] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Accepted: 02/25/2016] [Indexed: 11/22/2022] Open
Abstract
Asymmetric division (AD) is a fundamental mechanism whereby unequal inheritance of various cellular compounds during mitosis generates unequal fate in the two daughter cells. Unequal repartitions of transcription factors, receptors as well as mRNA have been abundantly described in AD. In contrast, the involvement of intermediate filaments in this process is still largely unknown. AD occurs in stem cells during development but was also recently observed in cancer stem cells. Here, we demonstrate the asymmetric distribution of the main astrocytic intermediate filament, namely the glial fibrillary acid protein (GFAP), in mitotic glioma multipotent cells isolated from glioblastoma (GBM), the most frequent type of brain tumor. Unequal mitotic repartition of GFAP was also observed in mice non-tumoral neural stem cells indicating that this process occurs across species and is not restricted to cancerous cells. Immunofluorescence and videomicroscopy were used to capture these rare and transient events. Considering the role of intermediate filaments in cytoplasm organization and cell signaling, we propose that asymmetric distribution of GFAP could possibly participate in the regulation of normal and cancerous neural stem cell fate.
Collapse
Affiliation(s)
- Pierre-Olivier Guichet
- INSERM U1051, Institut des Neurosciences de Montpellier, Hôpital St Eloi, 80 avenue Augustin Fliche, 34091 Montpellier Cedex 05, France
| | - Sophie Guelfi
- INSERM U1051, Institut des Neurosciences de Montpellier, Hôpital St Eloi, 80 avenue Augustin Fliche, 34091 Montpellier Cedex 05, France
| | - Chantal Ripoll
- INSERM U1051, Institut des Neurosciences de Montpellier, Hôpital St Eloi, 80 avenue Augustin Fliche, 34091 Montpellier Cedex 05, France
| | - Marisa Teigell
- INSERM U1051, Institut des Neurosciences de Montpellier, Hôpital St Eloi, 80 avenue Augustin Fliche, 34091 Montpellier Cedex 05, France
| | - Jean-Charles Sabourin
- INSERM U1051, Institut des Neurosciences de Montpellier, Hôpital St Eloi, 80 avenue Augustin Fliche, 34091 Montpellier Cedex 05, France
| | - Luc Bauchet
- CHU Montpellier, Hopital Gui de Chaulliac, 80, avenue Augustin Fliche, 34295 Montpellier, France
| | - Valérie Rigau
- CHU Montpellier, Hopital Gui de Chaulliac, 80, avenue Augustin Fliche, 34295 Montpellier, France
| | - Bernard Rothhut
- INSERM U1051, Institut des Neurosciences de Montpellier, Hôpital St Eloi, 80 avenue Augustin Fliche, 34091 Montpellier Cedex 05, France
| | - Jean-Philippe Hugnot
- INSERM U1051, Institut des Neurosciences de Montpellier, Hôpital St Eloi, 80 avenue Augustin Fliche, 34091 Montpellier Cedex 05, France
- Université Montpellier, Place Eugène Bataillon, 34095 Montpellier Cedex 5, France
- * E-mail: ;
| |
Collapse
|
9
|
Bhatt JM, Viktorova EG, Busby T, Wyrozumska P, Newman LE, Lin H, Lee E, Wright J, Belov GA, Kahn RA, Sztul E. Oligomerization of the Sec7 domain Arf guanine nucleotide exchange factor GBF1 is dispensable for Golgi localization and function but regulates degradation. Am J Physiol Cell Physiol 2015; 310:C456-69. [PMID: 26718629 DOI: 10.1152/ajpcell.00185.2015] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Accepted: 12/14/2015] [Indexed: 12/15/2022]
Abstract
Members of the large Sec7 domain-containing Arf guanine nucleotide exchange factor (GEF) family have been shown to dimerize through their NH2-terminal dimerization and cyclophilin binding (DCB) and homology upstream of Sec7 (HUS) domains. However, the importance of dimerization in GEF localization and function has not been assessed. We generated a GBF1 mutant (91/130) in which two residues required for oligomerization (K91 and E130 within the DCB domain) were replaced with A and assessed the effects of these mutations on GBF1 localization and cellular functions. We show that 91/130 is compromised in oligomerization but that it targets to the Golgi in a manner indistinguishable from wild-type GBF1 and that it rapidly exchanges between the cytosolic and membrane-bound pools. The 91/130 mutant appears active as it integrates within the functional network at the Golgi, supports Arf activation and COPI recruitment, and sustains Golgi homeostasis and cargo secretion when provided as a sole copy of functional GBF1 in cells. In addition, like wild-type GBF1, the 91/130 mutant supports poliovirus RNA replication, a process requiring GBF1 but believed to be independent of GBF1 catalytic activity. However, oligomerization appears to stabilize GBF1 in cells, and the 91/130 mutant is degraded faster than the wild-type GBF1. Our data support a model in which oligomerization is not a key regulator of GBF1 activity but impacts its function by regulating the cellular levels of GBF1.
Collapse
Affiliation(s)
- Jay M Bhatt
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Ekaterina G Viktorova
- Department of Veterinary Medicine, Virginia-Maryland Regional College of Veterinary Medicine, University of Maryland, College Park, Maryland; and
| | - Theodore Busby
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Paulina Wyrozumska
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Laura E Newman
- Department of Biochemistry, Emory University School of Medicine, Atlanta, Georgia
| | - Helen Lin
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Eunjoo Lee
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama
| | - John Wright
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama
| | - George A Belov
- Department of Veterinary Medicine, Virginia-Maryland Regional College of Veterinary Medicine, University of Maryland, College Park, Maryland; and
| | - Richard A Kahn
- Department of Biochemistry, Emory University School of Medicine, Atlanta, Georgia
| | - Elizabeth Sztul
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama;
| |
Collapse
|
10
|
Yu Z, Ge Y, Xie L, Zhang T, Huang L, Zhao X, Liu J, Huang G. Using a yeast two-hybrid system to identify FTCD as a new regulator for HIF-1α in HepG2 cells. Cell Signal 2014; 26:1560-6. [DOI: 10.1016/j.cellsig.2014.03.016] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2014] [Revised: 02/16/2014] [Accepted: 03/16/2014] [Indexed: 12/15/2022]
|
11
|
Wyrozumska P, Ashley JW, Ramanadham S, Liu Q, Garvey WT, Sztul E. Novel effects of Brefeldin A (BFA) in signaling through the insulin receptor (IR) pathway and regulating FoxO1-mediated transcription. CELLULAR LOGISTICS 2014; 4:e27732. [PMID: 24843827 PMCID: PMC4022606 DOI: 10.4161/cl.27732] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/14/2013] [Revised: 01/02/2014] [Accepted: 01/03/2014] [Indexed: 12/19/2022]
Abstract
Brefeldin A (BFA) is a fungal metabolite best known for its ability to inhibit activation of ADP-ribosylation factor (Arf) and thereby inhibit secretory traffic. BFA also appears to regulate the trafficking of the GLUT4 glucose transporter by inducing its relocation from intracellular stores to the cell surface. Such redistribution of GLUT4 is normally regulated by insulin-mediated signaling. Hence, we tested whether BFA may intersect with the insulin pathway. We report that BFA causes the activation of the insulin receptor (IR), IRS-1, Akt-2, and AS160 components of the insulin pathway. The response is mediated through phosphoinositol-3-kinase (PI3K) and Akt kinase since the PI3K inhibitor wortmannin and the Akt inhibitors MK2206 and perifosine inhibit the BFA effect. BFA-mediated activation of the insulin pathway results in Akt-mediated phosphorylation of the insulin-responsive transcription factor FoxO1. This leads to nuclear exclusion of FoxO1 and a decrease in transcription of the insulin-responsive gene SIRT-1. Our findings suggest novel effects for BFA in signaling and transcription, and imply that BFA has multiple intracellular targets and can be used to regulate diverse cellular responses that include vesicular trafficking, signaling and transcription.
Collapse
Affiliation(s)
- Paulina Wyrozumska
- Department of Cell, Developmental and Integrative Biology; University of Alabama at Birmingham; Birmingham, AL USA
| | - Jason W Ashley
- Department of Cell, Developmental and Integrative Biology; University of Alabama at Birmingham; Birmingham, AL USA
| | - Sasanka Ramanadham
- Department of Cell, Developmental and Integrative Biology; University of Alabama at Birmingham; Birmingham, AL USA
| | - Qinglan Liu
- Department of Nutrition Sciences University of Alabama at Birmingham; Birmingham, AL USA
| | - W Timothy Garvey
- Department of Nutrition Sciences University of Alabama at Birmingham; Birmingham, AL USA
| | - Elizabeth Sztul
- Department of Cell, Developmental and Integrative Biology; University of Alabama at Birmingham; Birmingham, AL USA
| |
Collapse
|
12
|
Delgui LR, Rodríguez JF, Colombo MI. The endosomal pathway and the Golgi complex are involved in the infectious bursal disease virus life cycle. J Virol 2013; 87:8993-9007. [PMID: 23741000 PMCID: PMC3754037 DOI: 10.1128/jvi.03152-12] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2012] [Accepted: 05/30/2013] [Indexed: 12/22/2022] Open
Abstract
Infectious bursal disease virus (IBDV), a double-stranded RNA virus belonging to the Birnaviridae family, causes immunosuppression in chickens. In this study, we defined the localization of IBDV replication complexes based on colocalization analysis of VP3, the major protein component of IBDV ribonucleoproteins (RNPs). Our results indicate that VP3 localizes to vesicular structures bearing features of early and late endocytic compartments located in the juxtanuclear region. Interfering with the endocytic pathway with a dominant negative version of Rab5 after the internalization step leads to a reduction in virus titer. Triple-immunostaining studies between VP3, the viral RNA-dependent RNA polymerase VP1, and viral double-stranded RNA (dsRNA) showed a well-defined colocalization, indicating that the three critical components of the RNPs colocalize in the same structure, likely representing replication complexes. Interestingly, recombinant expressed VP3 also localizes to endosomes. Employing Golgi markers, we found that VP3-containing vesicles were closely associated with this organelle. Depolymerization of microtubules with nocodazole caused a profound change in VP3 localization, showing a punctate distribution scattered throughout the cytoplasm. However, these VP3-positive structures remained associated with Golgi ministacks. Similarly, brefeldin A (BFA) treatment led to a punctate distribution of VP3, scattered throughout the cytoplasm of infected cells. In addition, analysis of intra- and extracellular viral infective particles after BFA treatment of avian cells suggested a role for the Golgi complex in viral assembly. These results constitute the first study elucidating the localization of IBDV replication complexes (i.e., in endocytic compartments) and establishing a role for the Golgi apparatus in the assembly step of a birnavirus.
Collapse
Affiliation(s)
- Laura R. Delgui
- Laboratorio de Biología Celular y Molecular, Instituto de Histología y Embriología de Mendoza, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo-CONICET, Mendoza, Argentina
- Instituto de Ciencias Básicas, Universidad Nacional de Cuyo, Mendoza, Argentina
| | - José F. Rodríguez
- Departamento de Biología Molecular y Celular, Centro Nacional de Biotecnología (CSIC), Madrid, Spain
| | - María I. Colombo
- Laboratorio de Biología Celular y Molecular, Instituto de Histología y Embriología de Mendoza, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo-CONICET, Mendoza, Argentina
| |
Collapse
|
13
|
Kelly C, Canning P, Buchanan PJ, Williams MT, Brown V, Gruenert DC, Elborn JS, Ennis M, Schock BC. Toll-like receptor 4 is not targeted to the lysosome in cystic fibrosis airway epithelial cells. Am J Physiol Lung Cell Mol Physiol 2013; 304:L371-82. [PMID: 23316065 PMCID: PMC4073939 DOI: 10.1152/ajplung.00372.2011] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2011] [Accepted: 01/03/2013] [Indexed: 01/13/2023] Open
Abstract
The innate immune response to bacterial infection is mediated through Toll-like receptors (TLRs), which trigger tightly regulated signaling cascades through transcription factors including NF-κB. LPS activation of TLR4 triggers internalization of the receptor-ligand complex which is directed toward lysosomal degradation or endocytic recycling. Cystic fibrosis (CF) patients display a robust and uncontrolled inflammatory response to bacterial infection, suggesting a defect in regulation. This study examined the intracellular trafficking of TLR4 in CF and non-CF airway epithelial cells following stimulation with LPS. We employed cells lines [16hBE14o-, CFBE41o- (CF), and CFTR-complemented CFBE41o-] and confirmed selected experiments in primary nasal epithelial cells from non-CF controls and CF patients (F508del homozygous). In control cells, TLR4 expression (surface and cytoplasmic) was reduced after LPS stimulation but remained unchanged in CF cells and was accompanied by a heightened inflammatory response 24 h after stimulation. All cells expressed markers of the early (EEA1) and late (Rab7b) endosomes at basal levels. However, only CF cells displayed persistent expression of Rab7b following LPS stimulation. Rab7 variants may directly internalize bacteria to the Golgi for recycling or to the lysosome for degradation. TLR4 colocalized with the lysosomal marker LAMP1 in 16 hBE14o- cells, suggesting that TLR4 is targeted for lysosomal degradation in these cells. However, this colocalization was not observed in CFBE41o- cells, where persistent expression of Rab7 and release of proinflammatory cytokines was detected. Consistent with the apparent inability of CF cells to target TLR4 toward the lysosome for degradation, we observed persistent surface and cytoplasmic expression of this pathogen recognition receptor. This defect may account for the prolonged cycle of chronic inflammation associated with CF.
Collapse
Affiliation(s)
- Catriona Kelly
- Centre for Infection and Immunity, Queen's University of Belfast, Belfast, UK
| | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Lowery J, Szul T, Styers M, Holloway Z, Oorschot V, Klumperman J, Sztul E. The Sec7 guanine nucleotide exchange factor GBF1 regulates membrane recruitment of BIG1 and BIG2 guanine nucleotide exchange factors to the trans-Golgi network (TGN). J Biol Chem 2013; 288:11532-45. [PMID: 23386609 PMCID: PMC3630886 DOI: 10.1074/jbc.m112.438481] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Three Sec7 guanine nucleotide exchange factors (GEFs) activate ADP-ribosylation factors (ARFs) to facilitate coating of transport vesicles within the secretory and endosomal pathways. GBF1 recruits COPI to pre-Golgi and Golgi compartments, whereas BIG1 and BIG2 recruit AP1 and GGA clathrin adaptors to the trans-Golgi network (TGN) and endosomes. Here, we report a functional cascade between these GEFs by showing that GBF1-activated ARFs (ARF4 and ARF5, but not ARF3) facilitate BIG1 and BIG2 recruitment to the TGN. We localize GBF1 ultrastructurally to the pre-Golgi, the Golgi, and also the TGN. Our findings suggest a model in which GBF1 localized within pre-Golgi and Golgi compartments mediates ARF activation to facilitate recruitment of COPI to membranes, whereas GBF1 localized at the TGN mediates ARF activation that leads to the recruitment of BIG1 and BIG2 to the TGN. Membrane-associated BIG1/2 then activates ARFs that recruit clathrin adaptors. In this cascade, an early acting GEF (GBF1) activates ARFs that mediate recruitment of late acting GEFs (BIG1/2) to coordinate coating events within the pre-Golgi/Golgi/TGN continuum. Such coordination may optimize the efficiency and/or selectivity of cargo trafficking through the compartments of the secretory pathway.
Collapse
Affiliation(s)
- Jason Lowery
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama 35294, USA
| | | | | | | | | | | | | |
Collapse
|
15
|
Cai C, Rajaram M, Zhou X, Liu Q, Marchica J, Li J, Powers RS. Activation of multiple cancer pathways and tumor maintenance function of the 3q amplified oncogene FNDC3B. Cell Cycle 2012; 11:1773-81. [PMID: 22510613 DOI: 10.4161/cc.20121] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
FNDC3B was recently identified in an oncogenomic screen for amplified oncogenes in hepatocellular carcinoma. It is located at 3q26 and is amplified in over 20% of cancers, usually as part of a broad amplified region encompassing the entire 3q arm. Consistent with an oncogenic role in multiple cancer types, we show here that overexpression of FNDC3B is capable of malignantly transforming mammary and kidney epithelial cells in addition to hepatocytes. To explore how FNDC3B transforms cells, we determined the cellular localization of its gene product and the cancer pathways that it activates. We found that the FNDC3B oncoprotein localizes to the Golgi network, and that its correct localization is essential for its transforming function. We found that overexpression of FNDC3B induces the epithelial-to-mesenchymal transition (EMT) and activates several cancer pathways, including PI3-kinase/Akt, Rb1 and TGFβ signaling. For TGFβ signaling, we analyzed the point in the pathway at which FNDC3B operates and obtained evidence that it induces expression of all three TGFβ ligands and also promotes TGFBR1 cell-surface localization. We found that RNAi-mediated knockdown of FNDC3B in cancer cells with 3q amplification suppressed their clonogenicity and tumorigenicity, but that the same RNAi knockdown had no effect on single-copy 3q cancer cells. These results indicate that FNDC3B is an important oncogenic driver gene of the 3q amplicon, adding to the growing list of oncogenic drivers within this commonly amplified region.
Collapse
Affiliation(s)
- Chunlin Cai
- Cancer Genome Center, Cold Spring Harbor Laboratory, Woodbury, NY, USA
| | | | | | | | | | | | | |
Collapse
|
16
|
Grabski R, Balklava Z, Wyrozumska P, Szul T, Brandon E, Alvarez C, Holloway ZG, Sztul E. Identification of a functional domain within the p115 tethering factor that is required for Golgi ribbon assembly and membrane trafficking. J Cell Sci 2012; 125:1896-909. [PMID: 22328511 DOI: 10.1242/jcs.090571] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022] Open
Abstract
The tethering factor p115 (known as Uso1p in yeast) has been shown to facilitate Golgi biogenesis and membrane traffic in cells in culture. However, the role of p115 within an intact animal is largely unknown. Here, we document that depletion of p115 by using RNA interference (RNAi) in C. elegans causes accumulation of the 170 kD soluble yolk protein (YP170) in the body cavity and retention of the yolk receptor RME-2 in the ER and the Golgi within oocytes. Structure-function analyses of p115 have identified two homology regions (H1 and H2) within the N-terminal globular head and the coiled-coil 1 (CC1) domain as essential for p115 function. We identify a new C-terminal domain of p115 as necessary for Golgi ribbon formation and cargo trafficking. We show that p115 mutants that lack the fourth CC domain (CC4) act in a dominant-negative manner to disrupt Golgi and prevent cargo trafficking in cells containing endogenous p115. Furthermore, using RNAi of p115 and the subsequent transfection with p115 deletion mutants, we show that CC4 is necessary for Golgi ribbon formation and membrane trafficking in cells depleted of endogenous p115. p115 has been shown to bind a subset of ER-Golgi SNAREs through CC1 and CC4 domains (Shorter et al., 2002). Our findings show that CC4 is required for p115 function, and suggest that both the CC1 and the CC4 SNARE-binding motifs participate in p115-mediated membrane tethering.
Collapse
Affiliation(s)
- Robert Grabski
- Department of Cell Biology, University of Alabama at Birmingham, Birmingham, AL 35924, USA
| | | | | | | | | | | | | | | |
Collapse
|
17
|
Groma G, Grskovic I, Schael S, Ehlen HWA, Wagener R, Fosang A, Aszodi A, Paulsson M, Brachvogel B, Zaucke F. Matrilin-4 is processed by ADAMTS-5 in late Golgi vesicles present in growth plate chondrocytes of defined differentiation state. Matrix Biol 2011; 30:275-80. [PMID: 21539915 DOI: 10.1016/j.matbio.2011.04.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2011] [Revised: 04/07/2011] [Accepted: 04/08/2011] [Indexed: 11/19/2022]
Abstract
The two aggrecanases ADAMTS-4 and ADAMTS-5 have been shown to not only play roles in the breakdown of cartilage extracellular matrix in osteoarthritis, but also mediate processing of matrilins in the secretory pathway. The matrilins are adaptor proteins with a function in connecting fibrillar and network-like components in the cartilage extracellular matrix. Cleavage resulting in processed matrilins with fewer ligand-binding subunits could make these less efficient in providing matrix cohesion. In this study, the processing and degradation of matrilin-4 during cartilage remodeling in the growth plate of the developing mouse long bones were studied in greater detail. We show that ADAMTS-5 and a matrilin-4 neoepitope, revealed upon ADAMTS cleavage, colocalize in prehypertrophic/hypertrophic chondrocytes while they are not detected in proliferating chondrocytes of the growth plate. ADAMTS-5 and the cleaved matrilin-4 are preferentially detected in vesicles derived from the Golgi apparatus. The matrilin-4 neoepitope was not observed in the growth plate of ADAMTS-5 deficient mice. We propose that in the growth plate ADAMTS-5, and not ADAMTS-4, has a physiological function in the intracellular processing of matrilins and potentially of other extracellular matrix proteins.
Collapse
Affiliation(s)
- Gergely Groma
- Center for Biochemistry, University of Cologne, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Harada K, Matsuoka H, Nakamura J, Fukuda M, Inoue M. Storage of GABA in chromaffin granules and not in synaptic-like microvesicles in rat adrenal medullary cells. J Neurochem 2010; 114:617-26. [DOI: 10.1111/j.1471-4159.2010.06792.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
19
|
Abstract
Background Membrane trafficking is a defining feature of eukaryotic cells, and is essential for the maintenance of organelle homeostasis and identity. We previously identified Scy1-like 1 (Scyl1), a member of the Scy1-like family of catalytically inactive protein kinases, as a high-affinity binding partner of COPI coats. COPI-coated vesicles control Golgi to endoplasmic reticulum trafficking and we observed that disruption of Scyl1 function leads to a decrease in trafficking of the KDEL receptor via the COPI pathway. We reasoned that if Scyl1 plays a major role in COPI trafficking its disruption could influence Golgi homeostasis. Methodology/Principal Findings We performed Scyl1 knock down in cultured cells using previously established methods and observed an alteration in Golgi morphology. Both the surface area and volume of the Golgi is increased in Scyl1-depleted cells, but the continuity and polarity of the organelle is unperturbed. At the ultrastructural level we observe a decrease in the orderly structure of the Golgi with an increase in cisternal luminal width, while the number of Golgi cisternae remains unchanged. The golgin family of proteins forms a detergent resistant network that controls Golgi homeostasis. Disruption of this protein network by knock down of the golgin p115 disrupts the Golgi localization of Scyl1. Moreover, we find that Scyl1 interacts with 58K/formiminotransferase cyclodeaminase (FTCD), a protein that is tightly associated with the cis face of the Golgi. Conclusions/Significance Our results place Scyl1 at an interface between the golgin network and COPI trafficking and demonstrate that Scyl1 is required for the maintenance of Golgi morphology. Coupled with the observation from others that Scyl1 is the gene product responsible for the neurodegenerative mouse model mdf, our results additionally implicate the regulation of COPI trafficking and Golgi homeostasis in neurodegeneration.
Collapse
Affiliation(s)
- Jonathon L. Burman
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | - Jason N. R. Hamlin
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | - Peter S. McPherson
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
- * E-mail:
| |
Collapse
|
20
|
Cell organization of the rat pars tuberalis. Evidence for open communication between pars tuberalis cells, cerebrospinal fluid and tanycytes. Cell Tissue Res 2009; 339:359-81. [DOI: 10.1007/s00441-009-0885-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2009] [Accepted: 09/09/2009] [Indexed: 10/20/2022]
|
21
|
Ushijima Y, Goshima F, Kimura H, Nishiyama Y. Herpes simplex virus type 2 tegument protein UL56 relocalizes ubiquitin ligase Nedd4 and has a role in transport and/or release of virions. Virol J 2009; 6:168. [PMID: 19835589 PMCID: PMC2770495 DOI: 10.1186/1743-422x-6-168] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2009] [Accepted: 10/16/2009] [Indexed: 12/26/2022] Open
Abstract
Background The ubiquitin system functions in a variety of cellular processes including protein turnover, protein sorting and trafficking. Many viruses exploit the cellular ubiquitin system to facilitate viral replication. In fact, herpes simplex virus (HSV) encodes a ubiquitin ligase (E3) and a de-ubiquitinating enzyme to modify the host's ubiquitin system. We have previously reported HSV type 2 (HSV-2) tegument protein UL56 as a putative adaptor protein of neuronal precursor cell-expressed developmentally down-regulated 4 (Nedd4) E3 ligase, which has been shown to be involved in protein sorting and trafficking. Results In this study, we visualized and characterized the dynamic intracellular localization of UL56 and Nedd4 using live-cell imaging and immunofluorescence analysis. UL56 was distributed to cytoplasmic vesicles, primarily to the trans-Golgi network (TGN), and trafficked actively throughout the cytoplasm. Moreover, UL56 relocalized Nedd4 to the vesicles in cells transiently expressing UL56 and in cells infected with HSV-2. We also investigated whether UL56 influenced the efficiency of viral replication, and found that extracellular infectious viruses were reduced in the absence of UL56. Conclusion These data suggest that UL56 regulates Nedd4 and functions to facilitate the cytoplasmic transport of virions from TGN to the plasma membrane and/or release of virions from the cell surface.
Collapse
Affiliation(s)
- Yoko Ushijima
- Department of Virology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan.
| | | | | | | |
Collapse
|
22
|
Calebiro D, Nikolaev VO, Gagliani MC, de Filippis T, Dees C, Tacchetti C, Persani L, Lohse MJ. Persistent cAMP-signals triggered by internalized G-protein-coupled receptors. PLoS Biol 2009; 7:e1000172. [PMID: 19688034 PMCID: PMC2718703 DOI: 10.1371/journal.pbio.1000172] [Citation(s) in RCA: 436] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2009] [Accepted: 07/07/2009] [Indexed: 01/19/2023] Open
Abstract
Real-time monitoring of G-protein-coupled receptor (GPCR) signaling in native cells suggests that the receptor for thyroid stimulating hormone remains active after internalization, challenging the current model for GPCR signaling. G-protein–coupled receptors (GPCRs) are generally thought to signal to second messengers like cyclic AMP (cAMP) from the cell surface and to become internalized upon repeated or prolonged stimulation. Once internalized, they are supposed to stop signaling to second messengers but may trigger nonclassical signals such as mitogen-activated protein kinase (MAPK) activation. Here, we show that a GPCR continues to stimulate cAMP production in a sustained manner after internalization. We generated transgenic mice with ubiquitous expression of a fluorescent sensor for cAMP and studied cAMP responses to thyroid-stimulating hormone (TSH) in native, 3-D thyroid follicles isolated from these mice. TSH stimulation caused internalization of the TSH receptors into a pre-Golgi compartment in close association with G-protein αs-subunits and adenylyl cyclase III. Receptors internalized together with TSH and produced downstream cellular responses that were distinct from those triggered by cell surface receptors. These data suggest that classical paradigms of GPCR signaling may need revision, as they indicate that cAMP signaling by GPCRs may occur both at the cell surface and from intracellular sites, but with different consequences for the cell. Cells respond to many environmental cues through the activity of cell surface receptor proteins, which sense these cues and convey that information to signaling molecules inside the cell. G-protein–coupled receptors (GPCRs) form the largest eukaryotic family of plasma membrane receptors. They convert the information provided by extracellular stimuli into intracellular second messengers, like cyclic AMP (cAMP). After prolonged stimulation, they are internalized inside cells, an event that to date has been thought to terminate the production of second messengers. Though many of the key steps of GPCR signaling are known in detail, precisely how signaling and termination actually occur in time and space (i.e., in subcellular compartments or microdomains) is still largely unexplored. To observe GPCR signaling in living cells, we generated mice expressing a fluorescent sensor that allows monitoring the intracellular levels of cAMP with a microscope. We utilized this system to study, directly in native thyroid follicles, the signal sent by the receptor for thyroid-stimulating hormone (TSH). Our findings indicate that TSH receptors are internalized rapidly after activation but continue to stimulate cAMP production inside cells and that this sustained, cAMP production is apparently required for localized activation of downstream components. These data challenge the current model of the GPCR-cAMP pathway by suggesting the existence of previously unrecognized intracellular site(s) for cAMP generation and of differential signaling outcomes as a result of intracellular GPCR signaling. Such intracellular site(s) may provide specialized signaling platforms, thus contributing to the spatiotemporal regulation of cAMP production and to signaling specificity within the GPCR family.
Collapse
Affiliation(s)
- Davide Calebiro
- Institute of Pharmacology and Toxicology, University of Würzburg, Würzburg, Germany
- Rudolf Virchow Center, DFG-Research Center for Experimental Biomedicine, University of Würzburg, Würzburg, Germany
- Dipartimento di Scienze Mediche, Università degli Studi di Milano, Milan, Italy
- Laboratory of Experimental Endocrinology, Fondazione IRCSS Istituto Auxologico Italiano, Cusano Milanino, Italy
- * E-mail: (DC); (MJL)
| | - Viacheslav O. Nikolaev
- Institute of Pharmacology and Toxicology, University of Würzburg, Würzburg, Germany
- Rudolf Virchow Center, DFG-Research Center for Experimental Biomedicine, University of Würzburg, Würzburg, Germany
| | | | - Tiziana de Filippis
- Laboratory of Experimental Endocrinology, Fondazione IRCSS Istituto Auxologico Italiano, Cusano Milanino, Italy
| | - Christian Dees
- Institute of Pharmacology and Toxicology, University of Würzburg, Würzburg, Germany
| | - Carlo Tacchetti
- Department of Experimental Medicine, University of Genoa, Genoa, Italy
| | - Luca Persani
- Dipartimento di Scienze Mediche, Università degli Studi di Milano, Milan, Italy
- Laboratory of Experimental Endocrinology, Fondazione IRCSS Istituto Auxologico Italiano, Cusano Milanino, Italy
| | - Martin J. Lohse
- Institute of Pharmacology and Toxicology, University of Würzburg, Würzburg, Germany
- Rudolf Virchow Center, DFG-Research Center for Experimental Biomedicine, University of Würzburg, Würzburg, Germany
- * E-mail: (DC); (MJL)
| |
Collapse
|
23
|
Kling P, Norman A, Andersson PL, Norrgren L, Förlin L. Gender-specific proteomic responses in zebrafish liver following exposure to a selected mixture of brominated flame retardants. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2008; 71:319-327. [PMID: 18258299 DOI: 10.1016/j.ecoenv.2007.12.010] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2007] [Revised: 12/05/2007] [Accepted: 12/08/2007] [Indexed: 05/25/2023]
Abstract
Proteomic effect screening in zebrafish liver was performed to generate hypotheses following exposure (21 days) to a structurally diverse mixture of brominated flame retardants (BFRs). Fish were exposed to two doses (10 and 100 nmol/g feed). Two-dimensional gel-electrophoresis, image analysis and MALDI-TOF mass-spectrometry revealed 13 and 19 significant responses in males and females, respectively. Effects on proteins related to cellular maintenance and stress were observed in both genders. Regulated proteins were gender-specific, but functionally indicated common protective responses (peroxiredoxin 6 and Zgc:92891 in males and transketolase in females) suggesting oxidative stress. Betaine homocysteine methyltransferase (BHMT) was induced in both genders. In addition a female-specific downregulation of ironhomeostatic proteins (iron-regulatory protein 1 and transferrin) were observed. Our proteomic approach revealed novel responses that suggest important gender-specific sensitivity to BFRs that should be considered when interpreting adverse effects of BFRs.
Collapse
Affiliation(s)
- P Kling
- Department of Zoology/Zoophysiology, Göteborg University, Box 463, SE-405 30 Göteborg, Sweden.
| | | | | | | | | |
Collapse
|
24
|
Burman JL, Bourbonniere L, Philie J, Stroh T, Dejgaard SY, Presley JF, McPherson PS. Scyl1, mutated in a recessive form of spinocerebellar neurodegeneration, regulates COPI-mediated retrograde traffic. J Biol Chem 2008; 283:22774-86. [PMID: 18556652 DOI: 10.1074/jbc.m801869200] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Scy1-like 1 (Scyl1), a member of the Scy1-like family of catalytically inactive protein kinases, was recently identified as the gene product altered in muscle-deficient mice, which suffer from motor neuron degeneration and cerebellar atrophy. To determine the function of Scyl1, we have now used a mass spectrometry-based screen to search for Scyl1-binding partners and identified components of coatomer I (COPI) coats. The interaction was confirmed in pull-down assays, and Scyl1 co-immunoprecipitates with betaCOP from brain lysates. Interestingly, and unique for a non-transmembrane domain protein, Scyl1 binds COPI coats using a C-terminal RKLD-COO(-) sequence, similar to the KKXX-COO(-) COPI-binding motif found in transmembrane endoplasmic reticulum (ER) proteins. Scyl1 co-localizes with betaCOP and is localized, in an Arf1-independent manner, to the ER-Golgi intermediate compartment and the cis-Golgi, sites of COPI-mediated membrane budding. The localization and binding properties of Scyl1 strongly suggest a function in COPI transport, and inhibitory RNA-mediated knock down of the protein disrupts COPI-mediated retrograde traffic of the KDEL receptor to the ER without affecting anterograde traffic from the ER. Our data demonstrate a function for Scyl1 as an accessory factor in COPI trafficking and suggest for the first time that alterations in the COPI pathway result in neurodegenerative disease.
Collapse
Affiliation(s)
- Jonathon L Burman
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, QC H3A 2B4, Canada
| | | | | | | | | | | | | |
Collapse
|
25
|
Styers ML, O'Connor AK, Grabski R, Cormet-Boyaka E, Sztul E. Depletion of beta-COP reveals a role for COP-I in compartmentalization of secretory compartments and in biosynthetic transport of caveolin-1. Am J Physiol Cell Physiol 2008; 294:C1485-98. [PMID: 18385291 DOI: 10.1152/ajpcell.00010.2008] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
We have utilized small interfering RNA (siRNA)-mediated depletion of the beta-COP subunit of COP-I to explore COP-I function in organellar compartmentalization and protein traffic. Reduction in beta-COP levels causes the colocalization of markers for the endoplasmic reticulum (ER)-Golgi intermediate compartment (ERGIC), Golgi, trans-Golgi network (TGN), and recycling endosomes in large, globular compartments. The lack of spatial differentiation of these compartments is not due to a general collapse of all cellular organelles since markers for the early endosomes and lysosomes do not redistribute to the common structures. Anterograde trafficking of the transmembrane cargo vesicular stomatitis virus membrane glycoprotein and of a subset of soluble cargoes is arrested within the common globular compartments. Similarly, recycling traffic of transferrin through the common compartment is perturbed. Furthermore, the trafficking of caveolin-1 (Cav1), a structural protein of caveolae, is arrested within the globular structures. Importantly, Cav1 coprecipitates with the gamma-subunit of COP-I, suggesting that Cav1 is a COP-I cargo. Our findings suggest that COP-I is required for the compartmentalization of the ERGIC, Golgi, TGN, and recycling endosomes and that COP-I plays a novel role in the biosynthetic transport of Cav1.
Collapse
Affiliation(s)
- Melanie L Styers
- Department of Cell Biology, University of Alabama at Birmingham, Birmingham, AL 35294, USa
| | | | | | | | | |
Collapse
|
26
|
Simultaneous tracking of capsid, tegument, and envelope protein localization in living cells infected with triply fluorescent herpes simplex virus 1. J Virol 2008; 82:5198-211. [PMID: 18353954 DOI: 10.1128/jvi.02681-07] [Citation(s) in RCA: 120] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
We report here the construction of a triply fluorescent-tagged herpes simplex virus 1 (HSV-1) expressing capsid protein VP26, tegument protein VP22, and envelope protein gB as fusion proteins with monomeric yellow, red, and cyan fluorescent proteins, respectively. The recombinant virus enabled us to monitor the dynamics of these capsid, tegument, and envelope proteins simultaneously in the same live HSV-1-infected cells and to visualize single extracellular virions with three different fluorescent emissions. In Vero cells infected by the triply fluorescent virus, multiple cytoplasmic compartments were found to be induced close to the basal surfaces of the infected cells (the adhesion surfaces of the infected cells on the solid growth substrate). Major capsid, tegument, and envelope proteins accumulated and colocalized in the compartments, as did marker proteins for the trans-Golgi network (TGN) which has been implicated to be the site of HSV-1 secondary envelopment. Moreover, formation of the compartments was correlated with the dynamic redistribution of the TGN proteins induced by HSV-1 infection. These results suggest that HSV-1 infection causes redistribution of TGN membranes to form multiple cytoplasmic compartments, possibly for optimal secondary envelopment. This is the first real evidence for the assembly of all three types of herpesvirus proteins-capsid, tegument, and envelope membrane proteins-in TGN.
Collapse
|
27
|
Poon BK, Chen X, Lu M, Vyas NK, Quiocho FA, Wang Q, Ma J. Normal mode refinement of anisotropic thermal parameters for a supramolecular complex at 3.42-A crystallographic resolution. Proc Natl Acad Sci U S A 2007; 104:7869-74. [PMID: 17470791 PMCID: PMC1876539 DOI: 10.1073/pnas.0701204104] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2007] [Indexed: 11/18/2022] Open
Abstract
Here we report a normal-mode-based protocol for modeling anisotropic thermal motions of proteins in x-ray crystallographic refinement. The foundation for this protocol is a recently developed elastic normal mode analysis that produces much more accurate eigenvectors without the tip effect. The effectiveness of the procedure is demonstrated on the refinement of a 3.42-A structure of formiminotransferase cyclodeaminase, a 0.5-MDa homooctameric enzyme. Using an order of magnitude fewer adjustable thermal parameters than the conventional isotropic refinement, this protocol resulted in a decrease of the values of R(cryst) and R(free) and improvements of the density map. Several poorly resolved regions in the original isotropically refined structure became clearer so that missing side chains were fitted easily and mistraced backbone was corrected. Moreover, the distribution of anisotropic thermal ellipsoids revealed functionally important structure flexibility. This normal-mode-based refinement is an effective way of describing anisotropic thermal motions in x-ray structures and is particularly attractive for the refinement of very large and flexible supramolecular complexes at moderate resolutions.
Collapse
Affiliation(s)
- Billy K. Poon
- Department of Bioengineering, Rice University, Houston, TX 77005
| | - Xiaorui Chen
- Graduate Program of Structural and Computational Biology and Molecular Biophysics and
| | - Mingyang Lu
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, One Baylor Plaza, BCM-125, Houston, TX 77030
| | - Nand K. Vyas
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, One Baylor Plaza, BCM-125, Houston, TX 77030
| | - Florante A. Quiocho
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, One Baylor Plaza, BCM-125, Houston, TX 77030
| | - Qinghua Wang
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, One Baylor Plaza, BCM-125, Houston, TX 77030
| | - Jianpeng Ma
- Department of Bioengineering, Rice University, Houston, TX 77005
- Graduate Program of Structural and Computational Biology and Molecular Biophysics and
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, One Baylor Plaza, BCM-125, Houston, TX 77030
| |
Collapse
|
28
|
Woźniak MJ, Allan VJ. Cargo selection by specific kinesin light chain 1 isoforms. EMBO J 2006; 25:5457-68. [PMID: 17093494 PMCID: PMC1679764 DOI: 10.1038/sj.emboj.7601427] [Citation(s) in RCA: 72] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2006] [Accepted: 10/16/2006] [Indexed: 12/25/2022] Open
Abstract
Kinesin-1 drives the movement of diverse cargoes, and it has been proposed that specific kinesin light chain (KLC) isoforms target kinesin-1 to these different structures. Here, we test this hypothesis using two in vitro motility assays, which reconstitute the movement of rough endoplasmic reticulum (RER) and vesicles present in a Golgi membrane fraction. We generated GST-tagged fusion proteins of KLC1B and KLC1D that included the tetratricopeptide repeat domain and the variable C-terminus. We find that preincubation of RER with KLC1B inhibits RER motility, whereas KLC1D does not. In contrast, Golgi fraction vesicle movement is inhibited by KLC1D but not KLC1B reagents. Both RER and vesicle movement is inhibited by preincubation with the GST-tagged C-terminal domain of ubiquitous kinesin heavy chain (uKHC), which binds to the N-terminal domain of uKHC and alters its interaction with microtubules. We propose that although the TRR domains are required for cargo binding, it is the variable C-terminal region of KLCs that are vital for targeting kinesin-1 to different cellular structures.
Collapse
Affiliation(s)
- Marcin J Woźniak
- Faculty of Life Sciences, University of Manchester, Manchester, UK
| | - Victoria J Allan
- Faculty of Life Sciences, University of Manchester, Manchester, UK
- Faculty of Life Sciences, University of Manchester, The Michael Smith Building, Oxford Road, Manchester M13 9PT, UK. Tel.: +44 161 275 5646; Fax: +44 161 275 5082; E-mail:
| |
Collapse
|
29
|
De Marco N, Buono M, Troise F, Diez-Roux G. Optineurin increases cell survival and translocates to the nucleus in a Rab8-dependent manner upon an apoptotic stimulus. J Biol Chem 2006; 281:16147-56. [PMID: 16569640 DOI: 10.1074/jbc.m601467200] [Citation(s) in RCA: 91] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
In glaucoma the retinal ganglion cells of the retina die through the induction of apoptosis leading to excavation of the optic nerve and blindness. Mutations in the optineurin (optic neuropathy inducing) protein were found associated with an adult form of glaucoma. To date, the role of optineurin in the neurodegeneration process that occurs during glaucoma is still unknown. We now report that in response to an apoptotic stimulus, optineurin changes subcellular localization and translocates from the Golgi to the nucleus. This translocation is dependent on the GTPase activity of Rab8, an interactor of optineurin. Furthermore, we demonstrate that the overexpression of optineurin protects cells from H2O2-induced cell death and blocks cytochrome c release from the mitochondria. A mutated form of optineurin, E50K, identified in normal tension glaucoma patients loses its ability to translocate to the nucleus and when overexpressed compromises the mitochondrial membrane integrity resulting in cells that are less fit to survive under stress conditions. The correlation between optineurin function and cell survival will be key to begin to understand retinal ganglion cell biology and signaling and to design general "survival" strategies to treat a disease of such a complex etiology as glaucoma.
Collapse
Affiliation(s)
- Nadia De Marco
- Telethon Institute of Genetics and Medicine, Via Pietro Castellino 111, 80131 Napoli, Italy
| | | | | | | |
Collapse
|
30
|
Hagiwara H, Tajika Y, Matsuzaki T, Suzuki T, Aoki T, Takata K. Localization of Golgi 58K protein (formiminotransferase cyclodeaminase) to the centrosome. Histochem Cell Biol 2006; 126:251-9. [PMID: 16534631 DOI: 10.1007/s00418-006-0166-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/19/2006] [Indexed: 10/24/2022]
Abstract
In vertebrate cells, the centrosome consists of a pair of centrioles and surrounding pericentriolar material. Using anti-Golgi 58K protein antibodies that recognize formiminotransferase cyclodeaminase (FTCD), we investigated its localization to the centrosome in various cultured cells and human oviductal secretory cells by immunohistochemistry. In addition to the Golgi apparatus, FTCD was localized to the centrosome, more abundantly around the mother centriole. The centrosome localization of FTCD continued throughout the cell cycle and was not disrupted after Golgi fragmentation, which was induced by colcemid and brefeldin A. Centriole microtubules are polyglutamylated and stable against tubulin depolymerizing drugs. FTCD in the centrosome may be associated with polyglutamylated residues of centriole microtubules and may play a role in providing centrioles with glutamate produced by cyclodeaminase domains of FTCD.
Collapse
Affiliation(s)
- Haruo Hagiwara
- Department of Anatomy and Cell Biology, Gunma University Graduate School of Medicine, 3-39-22, Showa-machi, Maebashi, Gunma 371-8511, Japan.
| | | | | | | | | | | |
Collapse
|
31
|
Abstract
Coiled-coil and multisubunit tethers have emerged as key regulators of membrane traffic and organellar architecture. The restricted subcellular localization of tethers and their ability to interact with Rabs and soluble N-ethylmaleimide-sensitive factor attachment protein receptors (SNAREs) suggests that tethers participate in determining the specificity of membrane fusion. An accepted model of tether function considers them molecular “bridges” that link opposing membranes before SNARE pairing. This model has been extended by findings in various experimental systems, suggesting that tethers may have other functions. Recent reports implicate tethers in the assembly of SNARE complexes, cargo selection and transit, cytoskeletal events, and localized attachment of regulatory proteins. A concept of tethers as scaffolding machines that recruit protein components involved in varied cellular responses is emerging. In this model, tethers function as integration switches that simultaneously transmit information to coordinate distinct processes required for membrane traffic.
Collapse
Affiliation(s)
- Elizabeth Sztul
- Dept. of Cell Biology, Univ. of Alabama at Birmingham, 1918 Univ. Blvd., Birmingham, AL 35294, USA.
| | | |
Collapse
|
32
|
Dresbach T, Torres V, Wittenmayer N, Altrock WD, Zamorano P, Zuschratter W, Nawrotzki R, Ziv NE, Garner CC, Gundelfinger ED. Assembly of active zone precursor vesicles: obligatory trafficking of presynaptic cytomatrix proteins Bassoon and Piccolo via a trans-Golgi compartment. J Biol Chem 2005; 281:6038-47. [PMID: 16373352 DOI: 10.1074/jbc.m508784200] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Neurotransmitter release from presynaptic nerve terminals is restricted to specialized areas of the plasma membrane, so-called active zones. Active zones are characterized by a network of cytoplasmic scaffolding proteins involved in active zone generation and synaptic transmission. To analyze the modes of biogenesis of this cytomatrix, we asked how Bassoon and Piccolo, two prototypic active zone cytomatrix molecules, are delivered to nascent synapses. Although these proteins may be transported via vesicles, little is known about the importance of a vesicular pathway and about molecular determinants of cytomatrix molecule trafficking. We found that Bassoon and Piccolo co-localize with markers of the trans-Golgi network in cultured neurons. Impairing vesicle exit from the Golgi complex, either using brefeldin A, recombinant proteins, or a low temperature block, prevented transport of Bassoon out of the soma. Deleting a newly identified Golgi-binding region of Bassoon impaired subcellular targeting of recombinant Bassoon. Overexpressing this region to specifically block Golgi binding of the endogenous protein reduced the concentration of Bassoon at synapses. These results suggest that, during the period of bulk synaptogenesis, a primordial cytomatrix assembles in a trans-Golgi compartment. They further indicate that transport via Golgi-derived vesicles is essential for delivery of cytomatrix proteins to the synapse. Paradigmatically this establishes Golgi transit as an obligatory step for subcellular trafficking of distinct cytoplasmic scaffolding proteins.
Collapse
Affiliation(s)
- Thomas Dresbach
- Leibniz Institute for Neurobiology, Brenneckestrasse 6, D-39118 Magdeburg, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Jin C, Zhang Y, Zhu H, Ahmed K, Fu C, Yao X. Human Yip1A specifies the localization of Yif1 to the Golgi apparatus. Biochem Biophys Res Commun 2005; 334:16-22. [PMID: 15990086 DOI: 10.1016/j.bbrc.2005.06.051] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2005] [Accepted: 06/07/2005] [Indexed: 11/27/2022]
Abstract
Yip1p and Yif1p are essential for transport from ER to Golgi stack during the early secretory pathway in budding yeast. Here, we report the identification and characterization of human Yif1. Sequence analysis revealed that human Yif1 (HsYif1), like most of the other YIP1 protein family members, contains multiple transmembrane segments. Double immunofluorescence study revealed co-distribution of HsYif1 with Golgi marker such as GS27. To delineate the function of HsYif1, we conducted a yeast two-hybrid assay and identified an interaction between human HsYif1 and HsYip1A, a homolog of yeast Yip1. In addition, our immunoprecipitation pull-down assay validates the interaction between HsYif1 and HsYip1A. Moreover, our immunofluorescence study demonstrates the co-distribution of HsYif1 and HsYip1A. Significantly, over-expression of mutant HsYip1A-lacked cytosolic region disrupts the localization of HsYif1 to the Golgi, suggesting that HsYip1A specifies the localization of HsYif1 to the Golgi. Therefore, we conclude that human Yip1A interacts with and determines the localization of HsYif1 to the Golgi apparatus.
Collapse
Affiliation(s)
- Changjiang Jin
- Laboratory of Cell Dynamics, University of Science and Technology of China, Hefei 230027, China
| | | | | | | | | | | |
Collapse
|
34
|
Szul T, Garcia-Mata R, Brandon E, Shestopal S, Alvarez C, Sztul E. Dissection of membrane dynamics of the ARF-guanine nucleotide exchange factor GBF1. Traffic 2005; 6:374-85. [PMID: 15813748 DOI: 10.1111/j.1600-0854.2005.00282.x] [Citation(s) in RCA: 70] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
ADP-ribosylation factor (ARF)-facilitated recruitment of COP I to membranes is required for secretory traffic. The guanine nucleotide exchange factor GBF1 activates ARF and regulates ARF/COP I dynamics at the endoplasmic reticulum (ER)-Golgi interface. Like ARF and coatomer, GBF1 peripherally associates with membranes. ADP-ribosylation factor and coatomer have been shown to rapidly cycle between membranes and cytosol, but the membrane dynamics of GBF1 are unknown. Here, we used fluorescence recovery after photobleaching to characterize the behavior of GFP-tagged GBF1. We report that GBF1 rapidly cycles between membranes and the cytosol (t1/2 is approximately 17 +/- 1 seconds). GBF1 cycles faster than GFP-tagged ARF, suggesting that in each round of association/dissociation, GBF1 catalyzes a single event of ARF activation, and that the activated ARF remains on membrane after GBF1 dissociation. Using three different approaches [expression of an inactive (E794K) GBF1 mutant, expression of the ARF1 (T31N) mutant with decreased affinity for GTP and Brefeldin A treatment], we show that GBF1 is stabilized on membranes when in a complex with ARF-GDP. GBF1 dissociation from ARF and membranes is triggered by its catalytic activity, i.e. the displacement of GDP and the subsequent binding of GTP to ARF. Our findings imply that continuous cycles of recruitment and dissociation of GBF1 to membranes are required for sustained ARF activation and COP I recruitment that underlies ER-Golgi traffic.
Collapse
Affiliation(s)
- Tomasz Szul
- Department of Cell Biology, University of Alabama at Birmingham, Birmingham, AL 35924, USA
| | | | | | | | | | | |
Collapse
|
35
|
Lin CM, Chen HJ, Leung CL, Parry DAD, Liem RKH. Microtubule actin crosslinking factor 1b: a novel plakin that localizes to the Golgi complex. J Cell Sci 2005; 118:3727-38. [PMID: 16076900 DOI: 10.1242/jcs.02510] [Citation(s) in RCA: 75] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
MACF1 (microtubule actin crosslinking factor), also called ACF7 (actin crosslinking family 7) is a cytoskeletal linker protein that can associate with both actin filaments and microtubules. We have identified a novel alternatively spliced isoform of MACF1. We named this isoform MACF1b and renamed the original isoform MACF1a. MACF1b is identical to MACF1a, except that it has a region containing plakin (or plectin) repeats in the middle of the molecule. MACF1b is ubiquitously expressed in adult tissues with especially high levels in the lung. We studied the subcellular localization of MACF1b proteins in mammalian cell lines. In two lung cell lines, MACF1b was chiefly localized to the Golgi complex. Upon treatments that disrupt the Golgi complex, MACF1b redistributed into the cytosol, but remained co-localized with the dispersed Golgi ministacks. MACF1b proteins can be detected in the enriched Golgi fraction by western blotting. The domain of MACF1b that targets it to the Golgi was found at the N-terminal part of the region that contains the plakin repeats. Reducing the level of MACF1 proteins by small-interfering RNA resulted in the dispersal of the Golgi complex.
Collapse
Affiliation(s)
- Chung-Ming Lin
- Department of Pathology, Columbia University College of Physicians and Surgeons, New York, NY 10032, USA
| | | | | | | | | |
Collapse
|
36
|
Bayer M, Fischer J, Kremerskothen J, Ossendorf E, Matanis T, Konczal M, Weide T, Barnekow A. Identification and characterization of Iporin as a novel interaction partner for rab1. BMC Cell Biol 2005; 6:15. [PMID: 15796781 PMCID: PMC1079803 DOI: 10.1186/1471-2121-6-15] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2004] [Accepted: 03/29/2005] [Indexed: 11/30/2022] Open
Abstract
Background The small GTPase rab1a and its isoform rab1b are essential regulating components in the vesicle transport between the ER and the Golgi apparatus. Rab1 is thought to act as a molecular switch and can change between an active GTP-bound and an inactive GDP-bound conformation. To elucidate the function of rab1, several approaches have been established to isolate effector proteins, which interact with the activated conformation of rab1. To date p115, GM130, golgin-84 and MICAL have been identified as direct interacting partners. Together with rab1, these molecules are components of a protein complex, which mediates and regulates intracellular vesicle transport. Results Here, we report the characterization of Iporin, which is similar to KIAA0375 as a novel rab1-interacting protein. It was initially identified by yeast two-hybrid screening experiments with the active mutant of rab1b (rab1b Q67R) as bait. Iporin contains a SH3 domain and two polyproline stretches, which are known to play a role in protein/protein interactions. In addition, Iporin encloses a RUN domain, which seems to be a major part of the rab1binding domain (R1BD). Iporin is ubiquitously expressed and immunofluorescence staining displays a cytosolic punctual distribution. Interestingly, we also show that Iporin interacts with another rab1 interacting partner, the GM130 protein. Conclusion Our results demonstrate that Iporin is a potential new interacting partner of rab1. Iporin is different from already identified rab1 interacting proteins concerning protein structure and cellular localization. We conclude that Iporin might function as a link between the targeting of ER derived vesicles, triggered by the rab1 GTPase and a signaling pathway regulated by molecules containing SH3 and/or poly-proline regions. The characterization of this novel intermolecular relation could help to elucidate how vesicles find their way from ER to the Golgi apparatus.
Collapse
Affiliation(s)
- Michael Bayer
- UKM Muenster, Albert-Schweitzer-Str. 33, D-48149 Muenster, Germany
| | - Julia Fischer
- Department of Experimental Tumorbiology, University of Muenster, Badestr. 9, D-48149 Muenster, Germany
| | - Joachim Kremerskothen
- Department of Experimental Tumorbiology, University of Muenster, Badestr. 9, D-48149 Muenster, Germany
| | - Edith Ossendorf
- Department of Experimental Tumorbiology, University of Muenster, Badestr. 9, D-48149 Muenster, Germany
| | - Theodoros Matanis
- Department of Experimental Tumorbiology, University of Muenster, Badestr. 9, D-48149 Muenster, Germany
| | - Magdalena Konczal
- Department of Experimental Tumorbiology, University of Muenster, Badestr. 9, D-48149 Muenster, Germany
| | - Thomas Weide
- Cilian AG, Johann-Krane-Weg 42, D-48149 Muenster, Germany
| | - Angelika Barnekow
- Department of Experimental Tumorbiology, University of Muenster, Badestr. 9, D-48149 Muenster, Germany
| |
Collapse
|
37
|
Hanaki KI, Ohka S, Yamamoto K, Nomoto A, Yoshikura H. Oligo(dA-dT)-dependent signal amplification for the detection of proteins in cells. Biotechniques 2004; 36:856-60, 862-3. [PMID: 15152606 DOI: 10.2144/04365pt01] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
An ultrasensitive protein detection system in situ named the ImmunoAT-tailing method was developed. It consists of three elementary processes: (i) detection of a protein by a primary antibody and a biotinylated secondary antibody; (ii) linking of biotinylated 15-base oligo(dA-dT) to the biotinylated immunocomplex via streptavidin; and (iii) self-priming elongation of oligo(dA-dT) by the Klenow fragment, 3' to 5' exo-. After the elongation reaction in the presence of dATP, dTTP, and dye-labeled dUTP, the protein was labeled with a large number of the dye molecules. The poly(dA-dT) elongated without the labeled nucleotides was detected by 4',6-diamidino-2-phenylindole (DAPI) staining. By combining the different labelings, double staining was possible. This ImmunoAT-tailing method has a specificity and sensitivity higher than that of tyramide signal amplification.
Collapse
Affiliation(s)
- Ken-ichi Hanaki
- Japan Society for the Promotion of Science, University of Tokyo, Tokyo, Japan.
| | | | | | | | | |
Collapse
|
38
|
Mao Y, Vyas NK, Vyas MN, Chen DH, Ludtke SJ, Chiu W, Quiocho FA. Structure of the bifunctional and Golgi-associated formiminotransferase cyclodeaminase octamer. EMBO J 2004; 23:2963-71. [PMID: 15272307 PMCID: PMC514939 DOI: 10.1038/sj.emboj.7600327] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2004] [Accepted: 06/21/2004] [Indexed: 12/13/2022] Open
Abstract
Mammalian formiminotransferase cyclodeaminase (FTCD), a 0.5 million Dalton homo-octameric enzyme, plays important roles in coupling histidine catabolism with folate metabolism and integrating the Golgi complex with the vimentin intermediate filament cytoskeleton. It is also linked to two human diseases, autoimmune hepatitis and glutamate formiminotransferase deficiency. Determination of the FTCD structure by X-ray crystallography and electron cryomicroscopy revealed that the eight subunits, each composed of distinct FT and CD domains, are arranged like a square doughnut. A key finding indicates that coupling of three subunits governs the octamer-dependent sequential enzyme activities, including channeling of intermediate and conformational change. The structure further shed light on the molecular nature of two strong antigenic determinants of FTCD recognized by autoantibodies from patients with autoimmune hepatitis and on the binding of thin vimentin filaments to the FTCD octamer.
Collapse
Affiliation(s)
- Yuxin Mao
- Howard Hughes Medical Institute, Baylor College of Medicine, Houston, TX, USA
| | - Nand K Vyas
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Meenakshi N Vyas
- Howard Hughes Medical Institute, Baylor College of Medicine, Houston, TX, USA
| | - Dong-Hua Chen
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX, USA
- National Center for Macromolecular Imaging, Baylor College of Medicine, Houston, TX, USA
| | - Steven J Ludtke
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX, USA
- National Center for Macromolecular Imaging, Baylor College of Medicine, Houston, TX, USA
| | - Wah Chiu
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX, USA
- National Center for Macromolecular Imaging, Baylor College of Medicine, Houston, TX, USA
| | - Florante A Quiocho
- Howard Hughes Medical Institute, Baylor College of Medicine, Houston, TX, USA
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX, USA
- Howard Hughes Medical Institute, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA. Tel.: +1 713 798 6565; Fax: +1 713 798 8516; E-mail:
| |
Collapse
|
39
|
Kaiser F, Kaufmann SHE, Zerrahn J. IIGP, a member of the IFN inducible and microbial defense mediating 47 kDa GTPase family, interacts with the microtubule binding protein hook3. J Cell Sci 2004; 117:1747-56. [PMID: 15075236 DOI: 10.1242/jcs.01039] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Innate immunity against intracellular pathogens is critically determined by an as yet unknown interferon (IFN)-inducible mechanism exerted by members of the 47 kDa GTPase family. The association of IGTP and IIGP with membranous compartments, the endoplasmic reticulum and, in addition in case of IIGP, the Golgi, implicate these GTPases in intracellular membrane trafficking or processing. We identified the cytoplasmic linker molecule hook3 as an interactor for IIGP by yeast two-hybrid screening. The physical complex between these molecules was present in lysates of IFNγ-stimulated macrophages as demonstrated by co-immunoprecipitation. Only a minor subfraction of total cellular IIGP or hook3 was co-purified, indicating that this interaction is either transient and/or involves distinct subpopulations of the total cellular pools of these molecules. Binding of IIGP to hook3 depends on a GTP-bound conformation. Hook3 is a microtubule-binding protein which participates in the organization of the cis-Golgi compartment. Both proteins were detected in the Golgi-membrane-enriched fraction upon subcellular fractionation. Apart from the Golgi localization of both proteins, hook3 was detected in perinuclear regions in close spatial proximity to IIGP, associated with the endoplasmic reticulum. Our experiments identify hook3 as the first cooperation partner of a member of the 47 kDa GTPase protein family and indicate that hook3 links in an IFNγ-inducible fashion to cytoskeleton-based membrane trafficking.
Collapse
Affiliation(s)
- Frank Kaiser
- Department of Immunology, Max-Planck-Institute for Infection Biology, Schumannstrasse 21-22, 10117 Berlin, Germany
| | | | | |
Collapse
|
40
|
Grabski R, Szul T, Sasaki T, Timpl R, Mayne R, Hicks B, Sztul E. Mutations in COCH that result in non-syndromic autosomal dominant deafness (DFNA9) affect matrix deposition of cochlin. Hum Genet 2003; 113:406-16. [PMID: 12928864 DOI: 10.1007/s00439-003-0992-7] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2003] [Accepted: 06/10/2003] [Indexed: 10/26/2022]
Abstract
The COCH gene mutated in autosomal dominant sensorineural deafness (DFNA9) encodes cochlin, a major constituent of the inner ear extracellular matrix. Sequence analysis of cochlin from DFNA9 patients identified five distinct single-amino-acid mutations within a conserved region (the LCCL domain) of cochlin. To define the molecular basis of DFNA9, we have generated myc-tagged wild-type and mutant cochlins and explored their behavior in transient transfection systems. Western blotting of cell lysates and culture media indicates that wild-type and mutant cochlins are synthesized and secreted in similar amounts. Immunofluorescent staining confirms that all are detected within the endoplasmic reticulum and the Golgi complex of transfected cells. Our findings suggest that COCH mutations are unlikely to cause abnormalities in secretion and suggest that extracellular events might cause DFNA9 pathology. In agreement, we show that wild-type cochlin accumulates in extracellular deposits that closely parallel the matrix component fibronectin, whereas mutant cochlins vary in the amount and pattern of extracellular material. Whereas some mutants exhibit an almost normal deposition pattern, some show complete lack of deposition. Our results suggest that DFNA9 results from gene products that fail to integrate correctly into the extracellular matrix. The partial or complete penetrance of integration defects suggests that DFNA9 pathology may be caused by multiple molecular mechanisms, including compromised ability of cochlin to self-assemble or to form appropriate complexes with other matrix components.
Collapse
Affiliation(s)
- Robert Grabski
- Department of Cell Biology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | | | | | | | | | | | | |
Collapse
|
41
|
Nozawa N, Daikoku T, Koshizuka T, Yamauchi Y, Yoshikawa T, Nishiyama Y. Subcellular localization of herpes simplex virus type 1 UL51 protein and role of palmitoylation in Golgi apparatus targeting. J Virol 2003; 77:3204-16. [PMID: 12584344 PMCID: PMC149782 DOI: 10.1128/jvi.77.5.3204-3216.2003] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
The herpes simplex virus type 1 (HSV-1) UL51 gene products are virion-associated phosphoproteins with apparent molecular masses of 27, 29, and 30 kDa in HSV-1-infected cells. In this study, we have investigated the intracellular localization and distribution of UL51 protein both in infected cells and in transfected cells expressing only UL51. We found that this protein colocalized closely with Golgi marker proteins such as the Golgi-58K protein and GM130 in transfected cells expressing only UL51. However, in infected cells, the UL51 protein localized to the juxtanuclear region but only partially colocalized with the Golgi maker proteins. Mutant protein analysis revealed that the N-terminal 15 amino acid residues of the UL51 protein sufficed for this Golgi localization property. The UL51 protein redistributed on addition of brefeldin A. This was prevented by pretreatment with 2-deoxyglucose and sodium azide, which results in ATP depletion, but not by pretreatment with NaF and AlCl(3), which activates heterotrimeric G proteins. Moreover, we found that palmitoylation of the UL51 protein through the N-terminal cysteine at position 9 was necessary for its Golgi localization. Protease digestion analysis suggested that the UL51 protein localized on the cytoplasmic face of the membrane in UL51-transfected cells, while in infected cells it localized mainly to the inside of cytoplasmic vesicles and/or the viral envelope. Transmission immunoelectron microscopy revealed an association of UL51 protein-specific labeling with cytoplasmic virions and also with some membranous structure. We infer from these observations that internalization of UL51 protein into the cytoplasmic vesicle and/or virion may occur in association with viral envelopment in HSV-infected cells.
Collapse
Affiliation(s)
- Naoki Nozawa
- Laboratory of Virology, Research Institute for Disease Mechanism and Control, Nagoya University School of Medicine, Showa-ku, Nagoya 466-8550, Japan
| | | | | | | | | | | |
Collapse
|
42
|
Royo T, Martínez-González J, Vilahur G, Badimon L. Differential intracellular trafficking of von Willebrand factor (vWF) and vWF propeptide in porcine endothelial cells lacking Weibel-Palade bodies and in human endothelial cells. Atherosclerosis 2003; 167:55-63. [PMID: 12618268 DOI: 10.1016/s0021-9150(02)00393-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Von Willebrand factor (vWF) is an adhesive protein involved in primary haemostasis virtually absent in the thoracic aorta of swine, an animal model widely used in thrombosis and atherosclerosis. By RT-PCR analysis we show that porcine aortic endothelial cells (PAEC) express the vWF gene, although vWF mRNA levels were 8+/-0.8-fold (p<0.05) or 290+/-8.9-fold (p<0.0001) lower than those in porcine pulmonary artery EC (PPEC) or human aortic EC (HAEC), respectively. Although vWF was rare in the thoracic aorta of swine, vWF propeptide (vWFpp) was present in the endothelium of this artery and in both primary and passaged PAEC. In addition, vWFpp but not vWF was detected in PAEC by Western blot. In PAEC neither vWFpp nor P-selectin immunostaining depicted Weibel-Palade bodies (WPB)-like structures, and acute stimuli (alpha-thrombin or the calcium ionophore A23187) did not increase vWF secretion. vWFpp co-localized with a Golgi marker, that cycles between the stacked Golgi (SG fraction) and earlier compartments of the secretory pathway. Our results confirm that PAEC express very low levels of vWF mRNA and indicate that in these cells, that do not have WPB, vWF and vWFpp have divergent intracellular trafficking pathways.
Collapse
Affiliation(s)
- Teresita Royo
- Instituto de Investigación Cardiovascular de Barcelona, CSIC-ICCC-Hospital de la Santa Creu i Sant Pau, Avda. Sant Antoni Maria Claret # 167, Spain
| | | | | | | |
Collapse
|
43
|
Ito A, Koma YI, Sohda M, Watabe K, Nagano T, Misumi Y, Nojima H, Kitamura Y. Localization of the PP2A B56gamma regulatory subunit at the Golgi complex: possible role in vesicle transport and migration. THE AMERICAN JOURNAL OF PATHOLOGY 2003; 162:479-89. [PMID: 12547706 PMCID: PMC1851168 DOI: 10.1016/s0002-9440(10)63842-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
The BL6 subline was derived from the F10 line, which was derived from the B16 mouse melanoma cell line. BL6 cells are more invasive than F10 cells and differ genetically from F10 cells by an alteration of the gene encoding the B56gamma regulatory subunit of protein phosphatase 2A (PP2A). This alteration results in the transcription of mRNA encoding a truncated variant of the B56gamma1 isoform (Deltagamma1). When F10 cells were stained with a polyclonal antibody that recognizes three B56gamma isoforms, B56gamma1, B56gamma2, and B56gamma3, the immunofluorescent signals co-localized well with the cis-Golgi marker proteins. When BL6 cells were fractionated in a sucrose gradient, B56gamma1 and B56gamma2, but not B56gamma3, were present in the Golgi-enriched fraction. This fraction also contained the catalytic subunit of PP2A. FLAG-tagged Deltagamma1 preferentially localized to the trans-Golgi area rather than the cis-Golgi. This localization was the same as that of FLAG-tagged B56gamma1. NIH3T3 cells stably expressing Deltagamma1 transported a mutant viral protein from the endoplasmic reticulum to the plasma membrane much faster than wild-type cells. Their directional migration, as assessed by the advance of cells into a cell-free area, was also elevated. As Deltagamma1 reduces the activity of the B56gamma-containing PP2A holoenzymes, these results suggest that the normal holoenzymes suppress vesicle transport and that Deltagamma1 might increase the invasive ability of BL6 cells by activating Golgi function.
Collapse
Affiliation(s)
- Akihiko Ito
- Department of Pathology, Osaka University Medical School/Graduate School of Frontier Bioscience, Suita, Osaka, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Abstract
The molecular characterization of the autoreactivities associated with autoimmune liver disease will improve their diagnosis and enhance understanding of their pathogenic mechanisms. Surprisingly, little is known about the nature of the major autoreactivities associated with type 1 AIH, including homogeneous ANA and antibodies to microfilaments [3]. Type 1 AIH is, however, the prototype of autoimmune liver disease [103].
Collapse
Affiliation(s)
- Francesco B Bianchi
- Department of Internal Medicine, Cardioangiology, Hepatology, Alma Mater Studiorum, University of Bologna, Policlinico Sant'Orsola-Malpighi, via Massarenti, 9 40138 Bologna, Italy.
| | | | | |
Collapse
|
45
|
Gao YS, Vrielink A, MacKenzie R, Sztul E. A novel type of regulation of the vimentin intermediate filament cytoskeleton by a Golgi protein. Eur J Cell Biol 2002; 81:391-401. [PMID: 12160147 DOI: 10.1078/0171-9335-00260] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Whether the highly dynamic structure of the vimentin intermediate filament (IF) cytoskeleton responds to cues from cellular organelles, and what proteins might participate in such events is largely unknown. We have shown previously that the Golgi protein formiminotransferase cyclodeaminase (FTCD) binds to vimentin filaments in vivo and in vitro, and that overexpression of FTCD causes dramatic rearrangements of the vimentin IF cytoskeleton (Gao and Sztul, J. Cell Biol. 152, 877-894, 2001). Using real-time imaging, we now show that FTCD causes bundling of individual thinner vimentin filaments into fibers and that the bundling always originates at the Golgi. FTCD appears to be the molecular "glue" since FTCD cross-links vimentin filaments in vitro. To initiate the analysis of structural determinants required for FTCD function in vimentin dynamics, we used structure-based design to generate individual formiminotransferase (FT) and cyclodeaminase (CD) domains, and to produce an enzymatically inactive FTCD. We show that the intact octameric structure is required for FTCD binding to vimentin filaments and for promoting filament assembly, but that eliminating enzymatic activity does not affect FTCD effects on the vimentin cytoskeleton. Our findings indicate that the Golgi protein FTCD is a potent modulator of the vimentin IF cytoskeleton, and suggest that the Golgi might act as a reservoir for proteins that regulate cytoskeletal dynamics.
Collapse
Affiliation(s)
- Ya-Sheng Gao
- Department of Cell Biology, University of Alabama at Birmingham, 35294, USA
| | | | | | | |
Collapse
|
46
|
Graves TK, Patel S, Dannies PS, Hinkle PM. Misfolded growth hormone causes fragmentation of the Golgi apparatus and disrupts endoplasmic reticulum-to-Golgi traffic. J Cell Sci 2001; 114:3685-94. [PMID: 11707520 DOI: 10.1242/jcs.114.20.3685] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
In some individuals with autosomal dominant isolated growth hormone deficiency, one copy of growth hormone lacks amino acids 32-71 and is severely misfolded. We transfected COS7 cells with either wild-type human growth hormone or Δ32-71 growth hormone and investigated subcellular localization of growth hormone and other proteins. Δ32-71 growth hormone was retained in the endoplasmic reticulum, whereas wild-type hormone accumulated in the Golgi apparatus. When cells transfected with wild-type or Δ32-71 growth hormone were dually stained for growth hormone and the Golgi markers β-COP, membrin or 58K, wild-type growth hormone was colocalized with the Golgi markers, but β-COP, membrin and 58K immunoreactivity was highly dispersed or undetectable in cells expressing Δ32-71 growth hormone. Examination of α-tubulin immunostaining showed that the cytoplasmic microtubular arrangement was normal in cells expressing wild-type growth hormone, but microtubule-organizing centers were absent in nearly all cells expressing Δ32-71 growth hormone. To determine whether Δ32-71 growth hormone would alter trafficking of a plasma membrane protein, we cotransfected the cells with the thyrotropin-releasing hormone (TRH) receptor and either wild-type or Δ32-71 growth hormone. Cells expressing Δ32-71 growth hormone, unlike those expressing wild-type growth hormone, failed to show normal TRH receptor localization or binding. Expression of Δ32-71 growth hormone also disrupted the trafficking of two secretory proteins, prolactin and secreted alkaline phosphatase. Δ32-71 growth hormone only weakly elicited the unfolded protein response as indicated by induction of BiP mRNA. Pharmacological induction of the unfolded protein response partially prevented deletion mutant-induced Golgi fragmentation and partially restored normal TRH receptor trafficking. The ability of some misfolded proteins to block endoplasmic reticulum-to-Golgi traffic may explain their toxic effects on host cells and suggests possible strategies for therapeutic interventions.
Collapse
Affiliation(s)
- T K Graves
- Department of Pharmacology and Physiology, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA. Department of Pharmacology, Yale University School of Medicine, New Haven, CT 06510, USA
| | | | | | | |
Collapse
|
47
|
Rey O, Young SH, Cantrell D, Rozengurt E. Rapid protein kinase D translocation in response to G protein-coupled receptor activation. Dependence on protein kinase C. J Biol Chem 2001; 276:32616-26. [PMID: 11410587 DOI: 10.1074/jbc.m101649200] [Citation(s) in RCA: 90] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Protein kinase D (PKD)/protein kinase C (PKC) mu is a serine/threonine protein kinase that can be activated by physiological stimuli like growth factors, antigen-receptor engagement and G protein-coupled receptor (GPCR) agonists via a phosphorylation-dependent mechanism that requires PKC activity. In order to investigate the dynamic mechanisms associated with GPCR signaling, the intracellular translocation of a green fluorescent protein-tagged PKD was analyzed by real-time visualization in fibroblasts and epithelial cells stimulated with bombesin, a GPCR agonist. We found that bombesin induced a rapidly reversible plasma membrane translocation of green fluorescent protein-tagged PKD, an event that can be divided into two distinct mechanistic steps. The first step, which is exclusively mediated by the cysteine-rich domain in the N terminus of PKD, involved its translocation from the cytosol to the plasma membrane. The second step, i.e. the rapid reverse translocation of PKD from the plasma membrane to the cytosol, required its catalytic domain and surprisingly PKC activity. These findings provide evidence for a novel mechanism by which PKC coordinates the translocation and activation of PKD in response to bombesin-induced GPCR activation.
Collapse
Affiliation(s)
- O Rey
- Unit of Signal Transduction and Gastrointestinal Cancer, Department of Medicine, UCLA School of Medicine, Los Angeles, California 90095, USA
| | | | | | | |
Collapse
|
48
|
Ding W, Albrecht B, Luo R, Zhang W, Stanley JR, Newbound GC, Lairmore MD. Endoplasmic reticulum and cis-Golgi localization of human T-lymphotropic virus type 1 p12(I): association with calreticulin and calnexin. J Virol 2001; 75:7672-82. [PMID: 11462039 PMCID: PMC115002 DOI: 10.1128/jvi.75.16.7672-7682.2001] [Citation(s) in RCA: 74] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Human T-lymphotropic virus type 1 (HTLV-1) is a complex retrovirus encoding regulatory and accessory genes in four open reading frames (ORF I to IV) of the pX region. We have demonstrated an important role of pX ORF I expression, which encodes p12(I), in establishment of HTLV-1 infection in a rabbit model and for optimal viral infectivity in quiescent primary lymphocytes. These data indicated that p12(I) may enhance lymphocyte activation and thereby promote virus infection. To further define the role of p12(I) in cell activation, we characterized the subcellular localization of p12(I) in transfected 293T cells and HeLa-Tat cells by multiple methods, including immunofluorescence confocal microscopy, electron microscopy, and subcellular fractionation. Herein, we demonstrate that p12(I) accumulates in the endoplasmic reticulum (ER) and cis-Golgi apparatus. The location of p12(I) was unchanged following treatments with both cycloheximide (blocking de novo protein synthesis) and brefeldin A (disrupting ER-to-Golgi protein transport), indicating that the protein is retained in the ER and cis-Golgi. Moreover, using coimmunoprecipitation assays, we identify the direct binding of p12(I) with both calreticulin and calnexin, resident ER proteins which regulate calcium storage. Our results indicate that p12(I) directly binds key regulatory proteins involved in calcium-mediated cell signaling and suggest a role of p12(I) in the establishment of HTLV-1 infection by activation of host cells.
Collapse
Affiliation(s)
- W Ding
- Department of Veterinary Biosciences, The Arthur G. James Cancer Hospital and Solove Research Institute, The Ohio State University, Columbus, Ohio 43210, USA
| | | | | | | | | | | | | |
Collapse
|
49
|
Lyons RJ, Deane R, Lynch DK, Ye ZS, Sanderson GM, Eyre HJ, Sutherland GR, Daly RJ. Identification of a novel human tankyrase through its interaction with the adaptor protein Grb14. J Biol Chem 2001; 276:17172-80. [PMID: 11278563 DOI: 10.1074/jbc.m009756200] [Citation(s) in RCA: 91] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Tankyrase is an ankyrin repeat-containing poly(ADP-ribose) polymerase originally isolated as a binding partner for the telomeric protein TRF1, but recently identified as a mitogen-activated protein kinase substrate implicated in regulation of Golgi vesicle trafficking. In this study, a novel human tankyrase, designated tankyrase 2, was isolated in a yeast two-hybrid screen as a binding partner for the Src homology 2 domain-containing adaptor protein Grb14. Tankyrase 2 is a 130-kDa protein, which lacks the N-terminal histidine/proline/serine-rich region of tankyrase, but contains a corresponding ankyrin repeat region, sterile alpha motif module, and poly(ADP-ribose) polymerase homology domain. The TANKYRASE 2 gene localizes to chromosome 10q23.2 and is widely expressed, with mRNA transcripts particularly abundant in skeletal muscle and placenta. Upon subcellular fractionation, both Grb14 and tankyrase 2 associate with the low density microsome fraction, and association of these proteins in vivo can be detected by co-immunoprecipitation analysis. Deletion analyses implicate the N-terminal 110 amino acids of Grb14 and ankyrin repeats 10-19 of tankyrase 2 in mediating this interaction. This study supports a role for the tankyrases in cytoplasmic signal transduction pathways and suggests that vesicle trafficking may be involved in the subcellular localization or signaling function of Grb14.
Collapse
Affiliation(s)
- R J Lyons
- Cancer Research Program, Garvan Institute of Medical Research, St. Vincent's Hospital, Sydney, New South Wales 2010, Australia
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Walenta JH, Didier AJ, Liu X, Krämer H. The Golgi-associated hook3 protein is a member of a novel family of microtubule-binding proteins. J Cell Biol 2001; 152:923-34. [PMID: 11238449 PMCID: PMC2198811 DOI: 10.1083/jcb.152.5.923] [Citation(s) in RCA: 150] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Microtubules are central to the spatial organization of diverse membrane-trafficking systems. Here, we report that Hook proteins constitute a novel family of cytosolic coiled coil proteins that bind to organelles and to microtubules. The conserved NH(2)-terminal domains of Hook proteins mediate attachment to microtubules, whereas the more divergent COOH-terminal domains mediate the binding to organelles. Human Hook3 bound to Golgi membranes in vitro and was enriched in the cis-Golgi in vivo. Unlike other cis-Golgi-associated proteins, however, a large fraction of Hook3 maintained its juxtanuclear localization after Brefeldin A treatment, indicating a Golgi-independent mechanism for Hook3 localization. Because overexpression of Hook3 caused fragmentation of the Golgi complex, we propose that Hook3 participates in defining the architecture and localization of the mammalian Golgi complex.
Collapse
Affiliation(s)
- Jason H. Walenta
- Center for Basic Neuroscience and Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, Texas 75390
| | - Aaron J. Didier
- Center for Basic Neuroscience and Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, Texas 75390
| | - Xinran Liu
- Center for Basic Neuroscience and Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, Texas 75390
| | - Helmut Krämer
- Center for Basic Neuroscience and Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, Texas 75390
| |
Collapse
|