1
|
Saji T, Endo M, Okada Y, Minami Y, Nishita M. KIF1C facilitates retrograde transport of lysosomes through Hook3 and dynein. Commun Biol 2024; 7:1305. [PMID: 39394274 PMCID: PMC11470034 DOI: 10.1038/s42003-024-07023-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 10/07/2024] [Indexed: 10/13/2024] Open
Abstract
Lysosomes, crucial cellular organelles, undergo bidirectional transport along microtubules, mediated by motor proteins such as cytoplasmic dynein-1 (dynein) and various kinesins. While the kinesin-3 family member KIF1C is established in mediating anterograde vesicle transport, its role in lysosomal transport remains unclear. Our study reveals that KIF1C unexpectedly supports the retrograde transport of lysosomes, driven by dynein, and contributes to their perinuclear localization. Notably, while KIF1C facilitates this perinuclear positioning, its motor activity is not required and, instead, exerts an inhibitory effect on this process. Mechanistically, KIF1C facilitates this process by interacting with the dynein-activating adaptor Hook3, which associates with the lysosome-anchored protein RUFY3. This regulatory mechanism is critical for the efficient degradation of cargo in autophagic and endocytic pathways. Our findings identify an unconventional, non-motor role for KIF1C in activating dynein-driven lysosomal transport, expanding our understanding of its functional diversity in cellular trafficking.
Collapse
Affiliation(s)
- Takeshi Saji
- Department of Biochemistry, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Mitsuharu Endo
- Division of Cell Physiology, Department of Physiology and Cell Biology, Graduate School of Medicine, Kobe University, Kobe, Japan
| | - Yasushi Okada
- Department of Cell Biology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Laboratory for Cell Polarity Regulation, RIKEN Center for Biosystems Dynamics Research (BDR), Osaka, Japan
- Department of Physics, Graduate School of Science, The University of Tokyo, Tokyo, Japan
- Universal Biology Institute (UBI) and International Research Center for Neurointelligence (WPI-IRCN), The University of Tokyo, Tokyo, Japan
| | - Yasuhiro Minami
- Division of Cell Physiology, Department of Physiology and Cell Biology, Graduate School of Medicine, Kobe University, Kobe, Japan
| | - Michiru Nishita
- Department of Biochemistry, Fukushima Medical University School of Medicine, Fukushima, Japan.
| |
Collapse
|
2
|
Rajanala K, Wedegaertner PB. Gβγ signaling regulates microtubule-dependent control of Golgi integrity. Cell Signal 2023; 106:110630. [PMID: 36805843 PMCID: PMC10079639 DOI: 10.1016/j.cellsig.2023.110630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 02/09/2023] [Accepted: 02/13/2023] [Indexed: 02/18/2023]
Abstract
Gβγ subunits regulate several non-canonical functions at distinct intracellular organelles. Previous studies have shown that Gβγ signaling at the Golgi is necessary to mediate vesicular protein transport function and to regulate mitotic Golgi fragmentation. Disruption of Golgi structure also occurs in response to microtubule depolymerizing agents, such as nocodazole. In this study, we use siRNA against Gβ1/2 or specific Gγ subunits to deplete their expression, and show that their knockdown causes a significant reduction in nocodazole-induced Golgi fragmentation. We establish that knockdown of Gβγ or inhibition of Gβγ with gallein resulted in decreased activation of protein kinase D (PKD) in response to nocodazole treatment. We demonstrate that restricting the amount of free Gβγ available for signaling by either inhibiting Gαi activation using pertussis toxin or by knockdown of the non-GPCR GEF, Girdin/GIV protein, results in a substantial decrease in nocodazole-induced Golgi fragmentation and PKD phosphorylation. Our results also indicate that depletion of Gβγ or inhibition with gallein or pertussis toxin significantly reduces the microtubule disruption-dependent Golgi fragmentation phenotype observed in cells transfected with mutant SOD1, a major causative protein in familial amyotrophic lateral sclerosis (ALS). These results provide compelling evidence that Gβγ signaling is critical for the regulation of Golgi integrity.
Collapse
Affiliation(s)
- Kalpana Rajanala
- Department of Biochemistry and Molecular Biology, Thomas Jefferson University, Sidney Kimmel Medical College, Philadelphia, PA 19107, United States of America
| | - Philip B Wedegaertner
- Department of Biochemistry and Molecular Biology, Thomas Jefferson University, Sidney Kimmel Medical College, Philadelphia, PA 19107, United States of America.
| |
Collapse
|
3
|
Divergent Contribution of the Golgi Apparatus to Microtubule Organization in Related Cell Lines. Int J Mol Sci 2022; 23:ijms232416178. [PMID: 36555819 PMCID: PMC9782006 DOI: 10.3390/ijms232416178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 12/14/2022] [Accepted: 12/15/2022] [Indexed: 12/23/2022] Open
Abstract
Membrane trafficking in interphase animal cells is accomplished mostly along the microtubules. Microtubules are often organized radially by the microtubule-organizing center to coordinate intracellular transport. Along with the centrosome, the Golgi often serves as a microtubule-organizing center, capable of nucleating and retaining microtubules. Recent studies revealed the role of a special subset of Golgi-derived microtubules, which facilitates vesicular traffic from this central transport hub of the cell. However, proteins essential for microtubule organization onto the Golgi might be differentially expressed in different cell lines, while many potential participants remain undiscovered. In the current work, we analyzed the involvement of the Golgi complex in microtubule organization in related cell lines. We studied two cell lines, both originating from green monkey renal epithelium, and found that they relied either on the centrosome or on the Golgi as a main microtubule-organizing center. We demonstrated that the difference in their Golgi microtubule-organizing activity was not associated with the well-studied proteins, such as CAMSAP3, CLASP2, GCC185, and GMAP210, but revealed several potential candidates involved in this process.
Collapse
|
4
|
Wu Y, Ding Y, Zheng X, Liao K. The molecular chaperone Hsp90 maintains Golgi organization and vesicular trafficking by regulating microtubule stability. J Mol Cell Biol 2021; 12:448-461. [PMID: 31560394 PMCID: PMC7333477 DOI: 10.1093/jmcb/mjz093] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 07/05/2019] [Accepted: 09/03/2019] [Indexed: 11/14/2022] Open
Abstract
Hsp90 is an abundant and special molecular chaperone considered to be the regulator of many transcription factors and signaling kinases. Its high abundance is indicative of its involvement in some more fundamental processes. In this study, we provide evidence that Hsp90 is required for microtubule stabilization, Golgi organization, and vesicular trafficking. We showed that Hsp90 is bound to microtubule-associated protein 4 (MAP4), which is essential for maintaining microtubule acetylation and stabilization. Hsp90 depletion led to the decrease in MAP4, causing microtubule deacetylation and destabilization. Furthermore, in Hsp90-depleted cells, the Golgi apparatus was fragmented and anterograde vesicle trafficking was impaired, with phenotypes similar to those induced by silencing MAP4. These disruptive effects of Hsp90 depletion could be rescued by the expression of exogenous MAP4 or the treatment of trichostatin A that increases microtubule acetylation as well as stability. Thus, microtubule stability is an essential cellular event regulated by Hsp90.
Collapse
Affiliation(s)
- Yuan Wu
- Key Laboratory of Systems Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Yubo Ding
- Key Laboratory of Systems Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Xiudan Zheng
- Key Laboratory of Systems Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Kan Liao
- Key Laboratory of Systems Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| |
Collapse
|
5
|
Vallés AS, Tenconi PE, Luquez JM, Furland NE. The inhibition of microtubule dynamics instability alters lipid homeostasis in TM4 Sertoli cells. Toxicol Appl Pharmacol 2021; 426:115607. [PMID: 34089742 DOI: 10.1016/j.taap.2021.115607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Revised: 05/27/2021] [Accepted: 05/30/2021] [Indexed: 10/21/2022]
Abstract
Sertoli cells (SC) structurally support and transport nutrients to germ cells during spermatogenesis facilitated by an active cytoskeleton. Chemical perturbation of SC microtubule (MT) dynamics instability leads to premature germ cell exfoliation demonstrating that this process is essential for male fertility, yet the effects of MT damaging drugs on SC lipid metabolism have been less explored. The aim of this study was to advance our understanding of how adequate SC MT dynamicity is needed to finely tune lipid homeostasis. To elucidate the role of MT dynamics instability on the latter, we suppressed MT dynamicity by long-term exposures to 10 nM of nocodazole (NCZ) on TM4-SC cultures. Inhibition of MT dynamics instability affected the distribution of [3H] arachidonate on TM4-SC. Triacylglycerols (TAG) exhibited a higher proportion of the [3H] label, with significantly lower percentages in the mitochondrial phospholipid cardiolipin, and notably, also in phosphatidylethanolamine. A noteworthy and progressive accumulation of lipid droplets during the period of exposure to NCZ was accompanied by increased TAG levels but not cholesterol levels in TM4-SC. NCZ-exposed cells reduced their mitochondrial membrane potential and increased ROS production without triggering apoptosis, had a compromised autophagic flux, and lost their transferrin expression. Although SC morphology was preserved, the NCZ-exposed cells displayed alteration of the normal organization of microfilaments (f-actin) and intermediate filaments (vimentin). Our findings suggest that a preserved MT dynamicity is essential in the maintenance of lipid and fatty acids homeostasis in SC, and thus highlights a novel target in these cells for drugs that impair MT dynamicity.
Collapse
Affiliation(s)
- A S Vallés
- Instituto de Investigaciones Bioquıímicas de Bahía Blanca, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional del Sur (UNS), Bahía Blanca, Argentina.
| | - P E Tenconi
- Instituto de Investigaciones Bioquıímicas de Bahía Blanca, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional del Sur (UNS), Bahía Blanca, Argentina
| | - J M Luquez
- Instituto de Investigaciones Bioquıímicas de Bahía Blanca, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional del Sur (UNS), Bahía Blanca, Argentina
| | - N E Furland
- Instituto de Investigaciones Bioquıímicas de Bahía Blanca, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional del Sur (UNS), Bahía Blanca, Argentina
| |
Collapse
|
6
|
Wu Y, Zheng X, Ding Y, Zhou M, Wei Z, Liu T, Liao K. The molecular chaperone Hsp90α deficiency causes retinal degeneration by disrupting Golgi organization and vesicle transportation in photoreceptors. J Mol Cell Biol 2021; 12:216-229. [PMID: 31408169 PMCID: PMC7181719 DOI: 10.1093/jmcb/mjz048] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2018] [Revised: 03/01/2019] [Accepted: 04/28/2019] [Indexed: 11/14/2022] Open
Abstract
Heat shock protein 90 (Hsp90) is an abundant molecular chaperone with two isoforms, Hsp90α and Hsp90β. Hsp90β deficiency causes embryonic lethality, whereas Hsp90α deficiency causes few abnormities except male sterility. In this paper, we reported that Hsp90α was exclusively expressed in the retina, testis, and brain. Its deficiency caused retinitis pigmentosa (RP), a disease leading to blindness. In Hsp90α-deficient mice, the retina was deteriorated and the outer segment of photoreceptor was deformed. Immunofluorescence staining and electron microscopic analysis revealed disintegrated Golgi and aberrant intersegmental vesicle transportation in Hsp90α-deficient photoreceptors. Proteomic analysis identified microtubule-associated protein 1B (MAP1B) as an Hsp90α-associated protein in photoreceptors. Hspα deficiency increased degradation of MAP1B by inducing its ubiquitination, causing α-tubulin deacetylation and microtubule destabilization. Furthermore, the treatment of wild-type mice with 17-DMAG, an Hsp90 inhibitor of geldanamycin derivative, induced the same retinal degeneration as Hsp90α deficiency. Taken together, the microtubule destabilization could be the underlying reason for Hsp90α deficiency-induced RP.
Collapse
Affiliation(s)
- Yuan Wu
- Key Laboratory of Systems Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Xiudan Zheng
- Key Laboratory of Systems Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Yubo Ding
- Key Laboratory of Systems Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Min Zhou
- Department of Ophthalmology, Eye and ENT Hospital of Fudan University, Shanghai 200031, China
| | - Zhuang Wei
- Key Laboratory of Systems Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Tao Liu
- Key Laboratory of Systems Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Kan Liao
- Key Laboratory of Systems Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| |
Collapse
|
7
|
The Chlamydia effector CT622/TaiP targets a nonautophagy related function of ATG16L1. Proc Natl Acad Sci U S A 2020; 117:26784-26794. [PMID: 33055216 DOI: 10.1073/pnas.2005389117] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The obligate intracellular bacteria Chlamydia trachomatis, the causative agent of trachoma and sexually transmitted diseases, multiply in a vacuolar compartment, the inclusion. From this niche, they secrete "effector" proteins, that modify cellular activities to enable bacterial survival and proliferation. Here, we show that the host autophagy-related protein 16-1 (ATG16L1) restricts inclusion growth and that this effect is counteracted by the secretion of the bacterial effector CT622/TaiP (translocated ATG16L1 interacting protein). ATG16L1 is mostly known for its role in the lipidation of the human homologs of ATG8 (i.e., LC3 and homologs) on double membranes during autophagy as well as on single membranes during LC3-associated phagocytosis and other LC3-lipidation events. Unexpectedly, the LC3-lipidation-related functions of ATG16L1 are not required for restricting inclusion development. We show that the carboxyl-terminal domain of TaiP exposes a mimic of an eukaryotic ATG16L1-binding motif that binds to ATG16L1's WD40 domain. By doing so, TaiP prevents ATG16L1 interaction with the integral membrane protein TMEM59 and allows the rerouting of Rab6-positive compartments toward the inclusion. The discovery that one bacterial effector evolved to target ATG16L1's engagement in intracellular traffic rather than in LC3 lipidation brings this "secondary" activity of ATG16L1 in full light and emphasizes its importance for maintaining host cell homeostasis.
Collapse
|
8
|
Eichwald C, Ackermann M, Fraefel C. Mammalian orthoreovirus core protein μ2 reorganizes host microtubule-organizing center components. Virology 2020; 549:13-24. [PMID: 32805585 PMCID: PMC7402380 DOI: 10.1016/j.virol.2020.07.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 07/02/2020] [Accepted: 07/08/2020] [Indexed: 10/26/2022]
Abstract
Filamentous mammalian orthoreovirus (MRV) viral factories (VFs) are membrane-less cytosolic inclusions in which virus transcription, replication of dsRNA genome segments, and packaging of virus progeny into newly synthesized virus cores take place. In infected cells, the MRV μ2 protein forms punctae in the enlarged region of the filamentous VFs that are co-localized with γ-tubulin and resistant to nocodazole treatment, and permitted microtubule (MT)-extension, features common to MT-organizing centers (MTOCs). Using a previously established reconstituted VF model, we addressed the functions of MT-components and MTOCs concerning their roles in the formation of filamentous VFs. Indeed, the MTOC markers γ-tubulin and centrin were redistributed within the VF-like structures (VFLS) in a μ2-dependent manner. Moreover, the MT-nucleation centers significantly increased in numbers, and γ-tubulin was pulled-down in a binding assay when co-expressed with histidine-tagged-μ2 and μNS. Thus, μ2, by interaction with γ-tubulin, can modulate MTOCs localization and function according to viral needs.
Collapse
Affiliation(s)
- Catherine Eichwald
- Institute of Virology, University of Zurich, Winterthurerstrasse 266a, 8057 Zurich, Switzerland.
| | - Mathias Ackermann
- Institute of Virology, University of Zurich, Winterthurerstrasse 266a, 8057 Zurich, Switzerland.
| | - Cornel Fraefel
- Institute of Virology, University of Zurich, Winterthurerstrasse 266a, 8057 Zurich, Switzerland.
| |
Collapse
|
9
|
Chin TM, Boopathy GTK, Man EP, Clohessy JG, Csizmadia E, Quinlan MP, Putti T, Wan SC, Xie C, Ali A, Wai FC, Ong YS, Goh BC, Settleman J, Hong W, Levantini E, Tenen DG. Targeting microtubules sensitizes drug resistant lung cancer cells to lysosomal pathway inhibitors. Am J Cancer Res 2020; 10:2727-2743. [PMID: 32194831 PMCID: PMC7052910 DOI: 10.7150/thno.38729] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Accepted: 11/24/2019] [Indexed: 12/20/2022] Open
Abstract
Oncogene-addicted cancers are predominantly driven by specific oncogenic pathways and display initial exquisite sensitivity to designer therapies, but eventually become refractory to treatments. Clear understanding of lung tumorigenic mechanisms is essential for improved therapies. Methods: Lysosomes were analyzed in EGFR-WT and mutant cells and corresponding patient samples using immunofluorescence or immunohistochemistry and immunoblotting. Microtubule organization and dynamics were studied using immunofluorescence analyses. Also, we have validated our findings in a transgenic mouse model that contain EGFR-TKI resistant mutations. Results: We herein describe a novel mechanism that a mutated kinase disrupts the microtubule organization and results in a defective endosomal/lysosomal pathway. This prevents the efficient degradation of phosphorylated proteins that become trapped within the endosomes and continue to signal, therefore amplifying downstream proliferative and survival pathways. Phenotypically, a distinctive subcellular appearance of LAMP1 secondary to microtubule dysfunction in cells expressing EGFR kinase mutants is seen, and this may have potential diagnostic applications for the detection of such mutants. We demonstrate that lysosomal-inhibitors re-sensitize resistant cells to EGFR tyrosine-kinase inhibitors (TKIs). Identifying the endosome-lysosome pathway and microtubule dysfunction as a mechanism of resistance allows to pharmacologically intervene on this pathway. Conclusions: We find that the combination of microtubule stabilizing agent and lysosome inhibitor could reduce the tumor progression in EGFR TKI resistant mouse models of lung cancer.
Collapse
|
10
|
Rodríguez-Cruz F, Torres-Cruz FM, Monroy-Ramírez HC, Escobar-Herrera J, Basurto-Islas G, Avila J, García-Sierra F. Fragmentation of the Golgi Apparatus in Neuroblastoma Cells Is Associated with Tau-Induced Ring-Shaped Microtubule Bundles. J Alzheimers Dis 2019; 65:1185-1207. [PMID: 30124450 DOI: 10.3233/jad-180547] [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] [Indexed: 12/26/2022]
Abstract
Abnormal fibrillary aggregation of tau protein is a pathological condition observed in Alzheimer's disease and other tauopathies; however, the presence and pathological significance of early non-fibrillary aggregates of tau remain under investigation. In cell and animal models expressing normal or modified tau, toxic effects altering the structure and function of several membranous organelles have also been reported in the absence of fibrillary structures; however, how these abnormalities are produced is an issue yet to be addressed. In order to obtain more insights into the mechanisms by which tau may disturb intracellular membranous elements, we transiently overexpressed human full-length tau and several truncated tau variants in cultured neuroblastoma cells. After 48 h of transfection, either full-length or truncated tau forms produced significant fragmentation of the Golgi apparatus (GA) with no changes in cell viability. Noteworthy is that in the majority of cells exhibiting dispersion of the GA, a ring-shaped array of cortical or perinuclear microtubule (Mt) bundles was also generated under the expression of either variant of tau. In contrast, Taxol treatment of non-transfected cells increased the amount of Mt bundles but not sufficiently to produce fragmentation of the GA. Tau-induced ring-shaped Mt bundles appeared to be well-organized and stable structures because they were resistant to Nocodazole post-treatment and displayed a high level of tubulin acetylation. These results further indicate that a mechanical force generated by tau-induced Mt-bundling may be responsible for Golgi fragmentation and that the repeated domain region of tau may be the main promoter of this effect.
Collapse
Affiliation(s)
- Fanny Rodríguez-Cruz
- Department of Cell Biology, Center of Research and Advanced Studies of the National Polytechnic Institute (CINVESTAV), Mexico City, Mexico
| | - Francisco Miguel Torres-Cruz
- Department of Cell Biology, Center of Research and Advanced Studies of the National Polytechnic Institute (CINVESTAV), Mexico City, Mexico
| | | | - Jaime Escobar-Herrera
- Department of Cell Biology, Center of Research and Advanced Studies of the National Polytechnic Institute (CINVESTAV), Mexico City, Mexico
| | | | - Jesús Avila
- Centro de Biología Molecular Severo Ochoa (CSIC-UAM) Universidad Autónoma de Madrid, Madrid, Spain
| | - Francisco García-Sierra
- Department of Cell Biology, Center of Research and Advanced Studies of the National Polytechnic Institute (CINVESTAV), Mexico City, Mexico
| |
Collapse
|
11
|
Tang CX, Luan L, Zhang L, Wang Y, Liu XF, Wang J, Xiong Y, Wang D, Huang LY, Gao DS. Golgin-160 and GMAP210 play an important role in U251 cells migration and invasion initiated by GDNF. PLoS One 2019; 14:e0211501. [PMID: 30695072 PMCID: PMC6351060 DOI: 10.1371/journal.pone.0211501] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Accepted: 01/15/2019] [Indexed: 11/25/2022] Open
Abstract
Gliomas are the most common malignant tumors of the brain and are characteristic of severe migration and invasion. Glial cell line-derived neurotrophic factor (GDNF) promotes glioma development process. However, the regulatory mechanisms of promoting occurrence and development of glioma have not yet been clearly elucidated. In the present study, the mechanism by which GDNF promotes glioma cell migration and invasion through regulating the dispersion and location of the Golgi apparatus (GA) is described. Following GDNF treatment, a change in the volume and position of GA was observed. The stack area of the GA was enlarged and it was more concentrated near the nucleus. Golgin-160 and Golgi microtubule-associated protein 210 (GMAP210) were identified as target molecules regulating GA positioning. In the absence of either golgin-160 or GMAP210 using lentivirus, the migration and invasion of U251 cells were decreased, while it was increased following GDNF. It was also found that the GA was decreased in size and dispersed following golgin-160 or GMAP210 knockdown, as determined by GA green fluorescence assay. Once GDNF was added, the above phenomenon would be twisted, and the concentrated location and volume of the GA was restored. In combination, the present data suggested that the regulation of the position and size of the GA by golgin-160 and GMAP210 play an important role in U251 cell migration and invasion.
Collapse
Affiliation(s)
- Chuan-Xi Tang
- Department of Neurobiology and Anatomy, Xuzhou Key Laboratory of Neurobiology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Lan Luan
- School of Nursing, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Lin Zhang
- School of Nursing, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yue Wang
- Department of Neurobiology and Anatomy, Xuzhou Key Laboratory of Neurobiology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Xin-Feng Liu
- Department of Neurosurgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Jie Wang
- Department of Neurobiology and Anatomy, Xuzhou Key Laboratory of Neurobiology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Ye Xiong
- Department of Neurobiology and Anatomy, Xuzhou Key Laboratory of Neurobiology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Dan Wang
- School of Medicine information, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Lin-Yan Huang
- School of Medical Technology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Dian-Shuai Gao
- Department of Neurobiology and Anatomy, Xuzhou Key Laboratory of Neurobiology, Xuzhou Medical University, Xuzhou, Jiangsu, China
- * E-mail:
| |
Collapse
|
12
|
Ringer K, Riehl J, Müller M, Dewes J, Hoff F, Jacob R. The large GTPase Mx1 binds Kif5B for cargo transport along microtubules. Traffic 2018; 19:947-964. [DOI: 10.1111/tra.12616] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Revised: 09/18/2018] [Accepted: 09/18/2018] [Indexed: 01/29/2023]
Affiliation(s)
- Karina Ringer
- Department of Cell Biology and Cell Pathology; Philipps University of Marburg; Marburg Germany
- DFG Research Training Group, Membrane Plasticity in Tissue Development and Remodeling; Philipps University of Marburg; Marburg Germany
| | - Jana Riehl
- Department of Cell Biology and Cell Pathology; Philipps University of Marburg; Marburg Germany
| | - Manuel Müller
- Department of Cell Biology and Cell Pathology; Philipps University of Marburg; Marburg Germany
| | - Jenny Dewes
- Department of Cell Biology and Cell Pathology; Philipps University of Marburg; Marburg Germany
| | - Florian Hoff
- Department of Cell Biology and Cell Pathology; Philipps University of Marburg; Marburg Germany
| | - Ralf Jacob
- Department of Cell Biology and Cell Pathology; Philipps University of Marburg; Marburg Germany
- DFG Research Training Group, Membrane Plasticity in Tissue Development and Remodeling; Philipps University of Marburg; Marburg Germany
| |
Collapse
|
13
|
Kalthur G, Salian SR, Nair R, Mathew J, Adiga SK, Kalthur SG, Zeegers D, Hande MP. Distribution pattern of cytoplasmic organelles, spindle integrity, oxidative stress, octamer-binding transcription factor 4 (Oct4) expression and developmental potential of oocytes following multiple superovulation. Reprod Fertil Dev 2018; 28:2027-2038. [PMID: 26173898 DOI: 10.1071/rd15184] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2015] [Accepted: 06/09/2015] [Indexed: 11/23/2022] Open
Abstract
The aim of the present study was to determine the effects of repeated superovulation on oocyte quality and embryo developmental potential. Female Swiss albino mice were injected with 5IU pregnant mare's serum gonadotropin followed 48h by 10IU human chorionic gonadotropin. Mice were superovulated up to four times with a gap of 7 days between each superovulation cycle. Ovarian weight increased significantly with an increasing number of superovulation cycles. Although the first stimulation cycle resulted in a threefold increase in the number of oocytes, the number of oocytes decreased gradually after subsequent stimulations. Increased cytoplasmic fragmentation, abnormal mitochondrial distribution, aggregation of Golgi apparatus, spindle damage, increased intracellular oxidative stress and a decrease in expression of octamer-binding transcription factor 4 (Oct4) expression were observed in these oocytes. Further, embryos derived from mice subjected to multiple stimulation cycles exhibited a low blastocyst rate, decreased hatching rate and increased apoptosis in blastocysts. In conclusion, the present study demonstrates that repeated superovulation adversely affects mouse oocyte quality by altering the distribution of cytoplasmic organelles, increasing oxidative stress and decreasing Oct4 expression, resulting in poor developmental potential of the embryos.
Collapse
Affiliation(s)
- Guruprasad Kalthur
- Division of Clinical Embryology, Department of Obstetrics and Gynecology, Kasturba Medical College, Manipal University, Manipal, 576104, India
| | - Sujith Raj Salian
- Division of Clinical Embryology, Department of Obstetrics and Gynecology, Kasturba Medical College, Manipal University, Manipal, 576104, India
| | - Ramya Nair
- Division of Clinical Embryology, Department of Obstetrics and Gynecology, Kasturba Medical College, Manipal University, Manipal, 576104, India
| | - Jemey Mathew
- Division of Clinical Embryology, Department of Obstetrics and Gynecology, Kasturba Medical College, Manipal University, Manipal, 576104, India
| | - Satish Kumar Adiga
- Division of Clinical Embryology, Department of Obstetrics and Gynecology, Kasturba Medical College, Manipal University, Manipal, 576104, India
| | | | - Dimphy Zeegers
- Genome Stability Laboratory, Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, 117597, Singapore
| | - M Prakash Hande
- Genome Stability Laboratory, Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, 117597, Singapore
| |
Collapse
|
14
|
She ZY, Pan MY, Tan FQ, Yang WX. Minus end-directed kinesin-14 KIFC1 regulates the positioning and architecture of the Golgi apparatus. Oncotarget 2018; 8:36469-36483. [PMID: 28430595 PMCID: PMC5482669 DOI: 10.18632/oncotarget.16863] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Accepted: 03/27/2017] [Indexed: 11/25/2022] Open
Abstract
The Golgi apparatus is the central organelle along the eukaryotic secretory and endocytic pathway. In non-polarized mammalian cells, the Golgi complex is usually located proximal to the nucleus at the cell center and is closely associated with the microtubule organizing center. Microtubule networks are essential in the organization and central localization of the Golgi apparatus, but the molecular basis underlying these processes are poorly understood. Here we reveal that minus end-directed kinesin-14 KIFC1 proteins are required for the structural integrity and positioning of the Golgi complex in non-polarized mammalian cells. Remarkably, we found that the motor domain of kinesin-14 KIFC1 regulates the recognition and binding of the Golgi and KIFC1 also statically binds to the microtubules via its tail domain. These findings reveal a new stationary binding model that kinesin-14 KIFC1 proteins function as crosslinkers between the Golgi apparatus and the microtubules and contribute to the central positioning and structural maintenance of the Golgi apparatus.
Collapse
Affiliation(s)
- Zhen-Yu She
- The Sperm Laboratory, College of Life Sciences, Zhejiang University, Hangzhou 310058, China
| | - Meng-Ying Pan
- The Sperm Laboratory, College of Life Sciences, Zhejiang University, Hangzhou 310058, China
| | - Fu-Qing Tan
- The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Wan-Xi Yang
- The Sperm Laboratory, College of Life Sciences, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
15
|
Land-locked mammalian Golgi reveals cargo transport between stable cisternae. Nat Commun 2017; 8:432. [PMID: 28874656 PMCID: PMC5585379 DOI: 10.1038/s41467-017-00570-z] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Accepted: 07/10/2017] [Indexed: 12/19/2022] Open
Abstract
The Golgi is composed of a stack of cis, medial, trans cisternae that are biochemically distinct. The stable compartments model postulates that permanent cisternae communicate through bi-directional vesicles, while the cisternal maturation model postulates that transient cisternae biochemically mature to ensure anterograde transport. Testing either model has been constrained by the diffraction limit of light microscopy, as the cisternae are only 10-20 nm thick and closely stacked in mammalian cells. We previously described the unstacking of Golgi by the ectopic adhesion of Golgi cisternae to mitochondria. Here, we show that cargo processing and transport continue-even when individual Golgi cisternae are separated and "land-locked" between mitochondria. With the increased spatial separation of cisternae, we show using three-dimensional live imaging that cis-Golgi and trans-Golgi remain stable in their composition and size. Hence, we provide new evidence in support of the stable compartments model in mammalian cells.The different composition of Golgi cisternae gave rise to two different models for intra-Golgi traffic: one where stable cisternae communicate via vesicles and another one where cisternae biochemically mature to ensure anterograde transport. Here, the authors provide evidence in support of the stable compartments model.
Collapse
|
16
|
Nishita M, Park SY, Nishio T, Kamizaki K, Wang Z, Tamada K, Takumi T, Hashimoto R, Otani H, Pazour GJ, Hsu VW, Minami Y. Ror2 signaling regulates Golgi structure and transport through IFT20 for tumor invasiveness. Sci Rep 2017; 7:1. [PMID: 28127051 PMCID: PMC5428335 DOI: 10.1038/s41598-016-0028-x] [Citation(s) in RCA: 8499] [Impact Index Per Article: 1062.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Accepted: 11/11/2016] [Indexed: 02/06/2023] Open
Abstract
Signaling through the Ror2 receptor tyrosine kinase promotes invadopodia formation for tumor invasion. Here, we identify intraflagellar transport 20 (IFT20) as a new target of this signaling in tumors that lack primary cilia, and find that IFT20 mediates the ability of Ror2 signaling to induce the invasiveness of these tumors. We also find that IFT20 regulates the nucleation of Golgi-derived microtubules by affecting the GM130-AKAP450 complex, which promotes Golgi ribbon formation in achieving polarized secretion for cell migration and invasion. Furthermore, IFT20 promotes the efficiency of transport through the Golgi complex. These findings shed new insights into how Ror2 signaling promotes tumor invasiveness, and also advance the understanding of how Golgi structure and transport can be regulated.
Collapse
Affiliation(s)
- Michiru Nishita
- Division of Cell Physiology, Department of Physiology and Cell Biology, Kobe University, Graduate School of Medicine, Kobe, 650-0017, Japan.
| | - Seung-Yeol Park
- Division of Rheumatology, Immunology and Allergy, Brigham and Women's Hospital, and Department of Medicine, Harvard Medical School, Boston, MA, 02115, USA
| | - Tadashi Nishio
- Division of Cell Physiology, Department of Physiology and Cell Biology, Kobe University, Graduate School of Medicine, Kobe, 650-0017, Japan
| | - Koki Kamizaki
- Division of Cell Physiology, Department of Physiology and Cell Biology, Kobe University, Graduate School of Medicine, Kobe, 650-0017, Japan
| | - ZhiChao Wang
- Division of Cell Physiology, Department of Physiology and Cell Biology, Kobe University, Graduate School of Medicine, Kobe, 650-0017, Japan
| | - Kota Tamada
- RIKEN Brain Science Institute, Wako, 351-0198, Japan
| | - Toru Takumi
- RIKEN Brain Science Institute, Wako, 351-0198, Japan
| | - Ryuju Hashimoto
- Department of Developmental Biology, Faculty of Medicine, Shimane University, Izumo, 690-8504, Japan
| | - Hiroki Otani
- Department of Developmental Biology, Faculty of Medicine, Shimane University, Izumo, 690-8504, Japan
| | - Gregory J Pazour
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA, 01605, USA
| | - Victor W Hsu
- Division of Rheumatology, Immunology and Allergy, Brigham and Women's Hospital, and Department of Medicine, Harvard Medical School, Boston, MA, 02115, USA
| | - Yasuhiro Minami
- Division of Cell Physiology, Department of Physiology and Cell Biology, Kobe University, Graduate School of Medicine, Kobe, 650-0017, Japan.
| |
Collapse
|
17
|
Xing M, Peterman MC, Davis RL, Oegema K, Shiau AK, Field SJ. GOLPH3 drives cell migration by promoting Golgi reorientation and directional trafficking to the leading edge. Mol Biol Cell 2016; 27:3828-3840. [PMID: 27708138 PMCID: PMC5170606 DOI: 10.1091/mbc.e16-01-0005] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Revised: 09/14/2016] [Accepted: 09/30/2016] [Indexed: 12/19/2022] Open
Abstract
The GOLPH3 oncogene functions in Golgi trafficking. GOLPH3 promotes cell migration, which is important in cancer. GOLPH3, by linking the Golgi to F-actin, promotes both Golgi reorientation and forward trafficking, which together drive trafficking to the leading edge. These findings provide insight into how GOLPH3 drives cell migration. The mechanism of directional cell migration remains an important problem, with relevance to cancer invasion and metastasis. GOLPH3 is a common oncogenic driver of human cancers, and is the first oncogene that functions at the Golgi in trafficking to the plasma membrane. Overexpression of GOLPH3 is reported to drive enhanced cell migration. Here we show that the phosphatidylinositol-4-phosphate/GOLPH3/myosin 18A/F-actin pathway that is critical for Golgi–to–plasma membrane trafficking is necessary and limiting for directional cell migration. By linking the Golgi to the actin cytoskeleton, GOLPH3 promotes reorientation of the Golgi toward the leading edge. GOLPH3 also promotes reorientation of lysosomes (but not other organelles) toward the leading edge. However, lysosome function is dispensable for migration and the GOLPH3 dependence of lysosome movement is indirect, via GOLPH3’s effect on the Golgi. By driving reorientation of the Golgi to the leading edge and driving forward trafficking, particularly to the leading edge, overexpression of GOLPH3 drives trafficking to the leading edge of the cell, which is functionally important for directional cell migration. Our identification of a novel pathway for Golgi reorientation controlled by GOLPH3 provides new insight into the mechanism of directional cell migration with important implications for understanding GOLPH3’s role in cancer.
Collapse
Affiliation(s)
- Mengke Xing
- Division of Endocrinology and Metabolism, Department of Medicine, University of California, San Diego, La Jolla, CA 92093
| | - Marshall C Peterman
- Division of Endocrinology and Metabolism, Department of Medicine, University of California, San Diego, La Jolla, CA 92093
| | - Robert L Davis
- Small Molecule Discovery Program, Ludwig Institute for Cancer Research, La Jolla, CA 92093
| | - Karen Oegema
- Department of Cellular and Molecular Medicine, Ludwig Institute for Cancer Research, University of California, San Diego, La Jolla, CA 92093
| | - Andrew K Shiau
- Small Molecule Discovery Program, Ludwig Institute for Cancer Research, La Jolla, CA 92093
| | - Seth J Field
- Division of Endocrinology and Metabolism, Department of Medicine, University of California, San Diego, La Jolla, CA 92093
| |
Collapse
|
18
|
Barker AR, McIntosh KV, Dawe HR. Centrosome positioning in non-dividing cells. PROTOPLASMA 2016; 253:1007-1021. [PMID: 26319517 DOI: 10.1007/s00709-015-0883-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2015] [Accepted: 08/22/2015] [Indexed: 06/04/2023]
Abstract
Centrioles and centrosomes are found in almost all eukaryotic cells, where they are important for organising the microtubule cytoskeleton in both dividing and non-dividing cells. The spatial location of centrioles and centrosomes is tightly controlled and, in non-dividing cells, plays an important part in cell migration, ciliogenesis and immune cell functions. Here, we examine some of the ways that centrosomes are connected to other organelles and how this impacts on cilium formation, cell migration and immune cell function in metazoan cells.
Collapse
Affiliation(s)
- Amy R Barker
- Biosciences, College of Life and Environmental Sciences, University of Exeter, Stocker Road, Exeter, EX4 4QD, UK
- Centre for Microvascular Research, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, EC1M 6BQ, London
| | - Kate V McIntosh
- Biosciences, College of Life and Environmental Sciences, University of Exeter, Stocker Road, Exeter, EX4 4QD, UK
| | - Helen R Dawe
- Biosciences, College of Life and Environmental Sciences, University of Exeter, Stocker Road, Exeter, EX4 4QD, UK.
| |
Collapse
|
19
|
Bellouze S, Baillat G, Buttigieg D, de la Grange P, Rabouille C, Haase G. Stathmin 1/2-triggered microtubule loss mediates Golgi fragmentation in mutant SOD1 motor neurons. Mol Neurodegener 2016; 11:43. [PMID: 27277231 PMCID: PMC4899909 DOI: 10.1186/s13024-016-0111-6] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Accepted: 06/01/2016] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Pathological Golgi fragmentation represents a constant pre-clinical feature of many neurodegenerative diseases including amyotrophic lateral sclerosis (ALS) but its molecular mechanisms remain hitherto unclear. RESULTS Here, we show that the severe Golgi fragmentation in transgenic mutant SOD1(G85R) and SOD1(G93A) mouse motor neurons is associated with defective polymerization of Golgi-derived microtubules, loss of the COPI coat subunit β-COP, cytoplasmic dispersion of the Golgi tether GM130, strong accumulation of the ER-Golgi v-SNAREs GS15 and GS28 as well as tubular/vesicular Golgi fragmentation. Data mining, transcriptomic and protein analyses demonstrate that both SOD1 mutants cause early presymptomatic and rapidly progressive up-regulation of the microtubule-destabilizing proteins Stathmins 1 and 2. Remarkably, mutant SOD1-triggered Golgi fragmentation and Golgi SNARE accumulation are recapitulated by Stathmin 1/2 overexpression but completely rescued by Stathmin 1/2 knockdown or the microtubule-stabilizing drug Taxol. CONCLUSIONS We conclude that Stathmin-triggered microtubule destabilization mediates Golgi fragmentation in mutant SOD1-linked ALS and potentially also in related motor neuron diseases.
Collapse
Affiliation(s)
- Sarah Bellouze
- Institut de Neurosciences de la Timone, UMR 7289, Centre National de la Recherche Scientifique (CNRS) and Aix-Marseille Université, 27 bd Jean Moulin, 13005 Marseille, France
| | - Gilbert Baillat
- Institut de Neurosciences de la Timone, UMR 7289, Centre National de la Recherche Scientifique (CNRS) and Aix-Marseille Université, 27 bd Jean Moulin, 13005 Marseille, France
| | - Dorothée Buttigieg
- Institut de Neurosciences de la Timone, UMR 7289, Centre National de la Recherche Scientifique (CNRS) and Aix-Marseille Université, 27 bd Jean Moulin, 13005 Marseille, France
| | - Pierre de la Grange
- GenoSplice technology, iPEPS - ICM, Hôpital Pitié Salpêtrière, 47/83, bd de l'Hôpital, 75013 Paris, France
| | - Catherine Rabouille
- Department of Cell Biology, Hubrecht Institute of the KNAW & UMC Utrecht, Uppsalalaan 8, 3584 CT Utrecht, Netherlands
| | - Georg Haase
- Institut de Neurosciences de la Timone, UMR 7289, Centre National de la Recherche Scientifique (CNRS) and Aix-Marseille Université, 27 bd Jean Moulin, 13005 Marseille, France.
| |
Collapse
|
20
|
Haase G, Rabouille C. Golgi Fragmentation in ALS Motor Neurons. New Mechanisms Targeting Microtubules, Tethers, and Transport Vesicles. Front Neurosci 2015; 9:448. [PMID: 26696811 PMCID: PMC4672084 DOI: 10.3389/fnins.2015.00448] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Accepted: 11/13/2015] [Indexed: 12/12/2022] Open
Abstract
Pathological alterations of the Golgi apparatus, such as its fragmentation represent an early pre-clinical feature of many neurodegenerative diseases and have been widely studied in the motor neuron disease amyotrophic lateral sclerosis (ALS). Yet, the underlying molecular mechanisms have remained cryptic. In principle, Golgi fragmentation may result from defects in three major classes of proteins: structural Golgi proteins, cytoskeletal proteins and molecular motors, as well as proteins mediating transport to and through the Golgi. Here, we present the different mechanisms that may underlie Golgi fragmentation in animal and cellular models of ALS linked to mutations in SOD1, TARDBP (TDP-43), VAPB, and C9Orf72 and we propose a novel one based on findings in progressive motor neuronopathy (pmn) mice. These mice are mutated in the TBCE gene encoding the cis-Golgi localized tubulin-binding cofactor E, one of five chaperones that assist in tubulin folding and microtubule polymerization. Loss of TBCE leads to alterations in Golgi microtubules, which in turn impedes on the maintenance of the Golgi architecture. This is due to down-regulation of COPI coat components, dispersion of Golgi tethers and strong accumulation of ER-Golgi SNAREs. These effects are partially rescued by the GTPase ARF1 through recruitment of TBCE to the Golgi. We hypothesize that defects in COPI vesicles, microtubules and their interaction may also underlie Golgi fragmentation in human ALS linked to other mutations, spinal muscular atrophy (SMA), and related motor neuron diseases. We also discuss the functional relevance of pathological Golgi alterations, in particular their potential causative, contributory, or compensatory role in the degeneration of motor neuron cell bodies, axons and synapses.
Collapse
Affiliation(s)
- Georg Haase
- Centre National de la Recherche Scientifique and Aix-Marseille Université UMR 7289, Institut de Neurosciences de la Timone Marseille, France
| | - Catherine Rabouille
- The Department of Cell Biology, Hubrecht Institute of the Royal Netherlands Academy of Arts and Sciences and University Medical Center Utrecht Utrecht, Netherlands
| |
Collapse
|
21
|
ER trapping reveals Golgi enzymes continually revisit the ER through a recycling pathway that controls Golgi organization. Proc Natl Acad Sci U S A 2015; 112:E6752-61. [PMID: 26598700 DOI: 10.1073/pnas.1520957112] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Whether Golgi enzymes remain localized within the Golgi or constitutively cycle through the endoplasmic reticulum (ER) is unclear, yet is important for understanding Golgi dependence on the ER. Here, we demonstrate that the previously reported inefficient ER trapping of Golgi enzymes in a rapamycin-based assay results from an artifact involving an endogenous ER-localized 13-kD FK506 binding protein (FKBP13) competing with the FKBP12-tagged Golgi enzyme for binding to an FKBP-rapamycin binding domain (FRB)-tagged ER trap. When we express an FKBP12-tagged ER trap and FRB-tagged Golgi enzymes, conditions precluding such competition, the Golgi enzymes completely redistribute to the ER upon rapamycin treatment. A photoactivatable FRB-Golgi enzyme, highlighted only in the Golgi, likewise redistributes to the ER. These data establish Golgi enzymes constitutively cycle through the ER. Using our trapping scheme, we identify roles of rab6a and calcium-independent phospholipase A2 (iPLA2) in Golgi enzyme recycling, and show that retrograde transport of Golgi membrane underlies Golgi dispersal during microtubule depolymerization and mitosis.
Collapse
|
22
|
Copeland SJ, Thurston SF, Copeland JW. Actin- and microtubule-dependent regulation of Golgi morphology by FHDC1. Mol Biol Cell 2015; 27:260-76. [PMID: 26564798 PMCID: PMC4713130 DOI: 10.1091/mbc.e15-02-0070] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Accepted: 11/04/2015] [Indexed: 01/08/2023] Open
Abstract
The coordinated action of the actin and microtubule cytoskeletal networks is required for Golgi ribbon assembly. The novel formin FHDC1 accumulates on the Golgi-derived microtubule network, where it acts to regulate Golgi ribbon assembly in an actin- and microtubule-dependent manner. The Golgi apparatus is the central hub of intracellular trafficking and consists of tethered stacks of cis, medial, and trans cisternae. In mammalian cells, these cisternae are stitched together as a perinuclear Golgi ribbon, which is required for the establishment of cell polarity and normal subcellular organization. We previously identified FHDC1 (also known as INF1) as a unique microtubule-binding member of the formin family of cytoskeletal-remodeling proteins. We show here that endogenous FHDC1 regulates Golgi ribbon formation and has an apparent preferential association with the Golgi-derived microtubule network. Knockdown of FHDC1 expression results in defective Golgi assembly and suggests a role for FHDC1 in maintenance of the Golgi-derived microtubule network. Similarly, overexpression of FHDC1 induces dispersion of the Golgi ribbon into functional ministacks. This effect is independent of centrosome-derived microtubules and instead likely requires the interaction between the FHDC1 microtubule-binding domain and the Golgi-derived microtubule network. These effects also depend on the interaction between the FHDC1 FH2 domain and the actin cytoskeleton. Thus our results suggest that the coordination of actin and microtubule dynamics by FHDC1 is required for normal Golgi ribbon formation.
Collapse
Affiliation(s)
- Sarah J Copeland
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Susan F Thurston
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - John W Copeland
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| |
Collapse
|
23
|
Egerer J, Emmerich D, Fischer-Zirnsak B, Chan WL, Meierhofer D, Tuysuz B, Marschner K, Sauer S, Barr FA, Mundlos S, Kornak U. GORAB Missense Mutations Disrupt RAB6 and ARF5 Binding and Golgi Targeting. J Invest Dermatol 2015; 135:2368-2376. [PMID: 26000619 DOI: 10.1038/jid.2015.192] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2014] [Revised: 04/21/2015] [Accepted: 04/23/2015] [Indexed: 11/09/2022]
Abstract
Gerodermia osteodysplastica is a hereditary segmental progeroid disorder affecting skin, connective tissues, and bone that is caused by loss-of-function mutations in GORAB. The golgin, RAB6-interacting (GORAB) protein localizes to the Golgi apparatus and interacts with the small GTPase RAB6. In this study, we used different approaches to shed more light on the recruitment of GORAB to this compartment. We show that GORAB best colocalizes with trans-Golgi markers and is rapidly displaced upon Brefeldin A exposition, indicating a loose association with Golgi membranes. A yeast two-hybrid screening revealed a specific interaction with the small GTPase ADP-ribosylation factor (ARF5) in its active, GTP-bound form. ARF5 and RAB6 bind to GORAB via the same internal Golgi-targeting RAB6 and ARF5 binding (IGRAB) domain. Two GORAB missense mutations identified in gerodermia osteodysplastica patients fall within this IGRAB domain. GORAB carrying the mutation p.Ala220Pro had a cytoplasmic distribution and failed to interact with both RAB6 and ARF5. In contrast, the p.Ser175Phe mutation displaced GORAB from the Golgi compartment to vesicular structures and selectively impaired ARF5 binding. Our findings indicate that the IGRAB domain is crucial for the Golgi localization of GORAB and that loss of this localization impairs its physiological function.
Collapse
Affiliation(s)
- Johannes Egerer
- Institut fuer Medizinische Genetik und Humangenetik, Charité-Universitaetsmedizin Berlin, Berlin, Germany; Max-Planck-Institut fuer Molekulare Genetik, FG Development & Disease, Berlin, Germany
| | - Denise Emmerich
- Institut fuer Medizinische Genetik und Humangenetik, Charité-Universitaetsmedizin Berlin, Berlin, Germany; Max-Planck-Institut fuer Molekulare Genetik, FG Development & Disease, Berlin, Germany
| | - Björn Fischer-Zirnsak
- Institut fuer Medizinische Genetik und Humangenetik, Charité-Universitaetsmedizin Berlin, Berlin, Germany; Max-Planck-Institut fuer Molekulare Genetik, FG Development & Disease, Berlin, Germany
| | - Wing Lee Chan
- Institut fuer Medizinische Genetik und Humangenetik, Charité-Universitaetsmedizin Berlin, Berlin, Germany; Max-Planck-Institut fuer Molekulare Genetik, FG Development & Disease, Berlin, Germany
| | - David Meierhofer
- Max-Planck-Institut fuer Molekulare Genetik, Mass Spectrometry Facility, Berlin, Germany
| | - Beyhan Tuysuz
- Department of Pediatric Genetics, Cerrahpaşa Medical Faculty, Istanbul University, Istanbul, Turkey
| | - Katrin Marschner
- Institut fuer Medizinische Genetik und Humangenetik, Charité-Universitaetsmedizin Berlin, Berlin, Germany
| | - Sascha Sauer
- Max-Planck-Institut fuer Molekulare Genetik, Otto-Warburg-Laboratories, Berlin, Germany
| | - Francis A Barr
- Department of Biochemistry, University of Oxford, Oxford, UK
| | - Stefan Mundlos
- Institut fuer Medizinische Genetik und Humangenetik, Charité-Universitaetsmedizin Berlin, Berlin, Germany; Max-Planck-Institut fuer Molekulare Genetik, FG Development & Disease, Berlin, Germany; Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité-Universitaetsmedizin Berlin, Berlin, Germany
| | - Uwe Kornak
- Institut fuer Medizinische Genetik und Humangenetik, Charité-Universitaetsmedizin Berlin, Berlin, Germany; Max-Planck-Institut fuer Molekulare Genetik, FG Development & Disease, Berlin, Germany; Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité-Universitaetsmedizin Berlin, Berlin, Germany.
| |
Collapse
|
24
|
RhoD is a Golgi component with a role in anterograde protein transport from the ER to the plasma membrane. Exp Cell Res 2015; 333:208-219. [DOI: 10.1016/j.yexcr.2015.02.023] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2014] [Revised: 02/24/2015] [Accepted: 02/26/2015] [Indexed: 11/22/2022]
|
25
|
Zafar S, Schmitz M, Younus N, Tahir W, Shafiq M, Llorens F, Ferrer I, Andéoletti O, Zerr I. Creutzfeldt-Jakob Disease Subtype-Specific Regional and Temporal Regulation of ADP Ribosylation Factor-1-Dependent Rho/MLC Pathway at Pre-Clinical Stage. J Mol Neurosci 2015; 56:329-48. [PMID: 25896910 DOI: 10.1007/s12031-015-0544-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Accepted: 03/09/2015] [Indexed: 10/23/2022]
Abstract
Small GTPases of the Arf family mainly activate the formation of coated carrier vesicles. We showed that class-I Arf1 interacts specifically with full length GPI-anchored cellular prion protein (PrP(C)). Several recent reports have also demonstrated a missing link between the endoplasmic reticulum and the Golgi-complex role for proper folding, but the exact molecular mechanism is not yet fully understood. In the present study, we identified and characterized the interactive role of Arf1 during PrP(C) intracellular distribution under pathophysiological conditions. PrP(C) interaction with Arf1 was investigated in cortical primary neuronal cultures of PrP(C) wild type and knockout mice (PrP(-/-)). Arf1 and PrP(C) co-binding affinity was confirmed using reverse co-immunoprecipitation, co-localization affinity using confocal laser-scanning microscopy. Treatment with brefeldin-A modulated Arf1 expression and resulted in down-regulation and redistribution of PrP(C) into cytosolic region. In the pre-symptomatic stage of the disease, Arf1 expression was significantly downregulated in the frontal cortex in tg340 mice expressing about fourfold of human PrP-M129 with PrP null background that had been inoculated with human sCJD MM1 brain tissue homogenates (sCJD MM1 mice). In addition, the frontal cortex of CJD human brain demonstrated significant binding capacity of Arf1 protein using co-immunoprecipitation analysis. We also examined Arf1 expression in the brain of CJD patients with the subtypes MM1 and VV2 and found that it was regulated in a region-specific manner. In the frontal cortex, Arf1 expression was not significantly changed in either MM1 or VV2 subtype. Interestingly, Arf1 expression was significantly reduced in the cerebellum in both subtypes as compared to controls. Furthermore, we observed altered RhoA activity, which in turn affects myosin light-chain (MLC) phosphorylation and Arf1-dependent PI3K pathway. Together, our findings underscore a key early symptomatic role of Arf1 in neurodegeneration. Targeting the Arf/Rho/MLC signaling axis might be a promising strategy to uncover the missing link which probably influences disease progression and internal homeostasis of misfolded proteins.
Collapse
Affiliation(s)
- Saima Zafar
- Department of Neurology, Clinical Dementia Center and DZNE, Georg-August University, University Medical Center Goettingen (UMG), Robert-Koch-Str. 40, 37075, Goettingen, Germany,
| | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Harrington CR, Storey JMD, Clunas S, Harrington KA, Horsley D, Ishaq A, Kemp SJ, Larch CP, Marshall C, Nicoll SL, Rickard JE, Simpson M, Sinclair JP, Storey LJ, Wischik CM. Cellular Models of Aggregation-dependent Template-directed Proteolysis to Characterize Tau Aggregation Inhibitors for Treatment of Alzheimer Disease. J Biol Chem 2015; 290:10862-75. [PMID: 25759392 PMCID: PMC4409250 DOI: 10.1074/jbc.m114.616029] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2014] [Indexed: 12/15/2022] Open
Abstract
Alzheimer disease (AD) is a degenerative tauopathy characterized by aggregation of Tau protein through the repeat domain to form intraneuronal paired helical filaments (PHFs). We report two cell models in which we control the inherent toxicity of the core Tau fragment. These models demonstrate the properties of prion-like recruitment of full-length Tau into an aggregation pathway in which template-directed, endogenous truncation propagates aggregation through the core Tau binding domain. We use these in combination with dissolution of native PHFs to quantify the activity of Tau aggregation inhibitors (TAIs). We report the synthesis of novel stable crystalline leucomethylthioninium salts (LMTX®), which overcome the pharmacokinetic limitations of methylthioninium chloride. LMTX®, as either a dihydromesylate or a dihydrobromide salt, retains TAI activity in vitro and disrupts PHFs isolated from AD brain tissues at 0.16 μM. The Ki value for intracellular TAI activity, which we have been able to determine for the first time, is 0.12 μM. These values are close to the steady state trough brain concentration of methylthioninium ion (0.18 μM) that is required to arrest progression of AD on clinical and imaging end points and the minimum brain concentration (0.13 μM) required to reverse behavioral deficits and pathology in Tau transgenic mice.
Collapse
Affiliation(s)
- Charles R Harrington
- From the School of Medicine and Dentistry, University of Aberdeen, Aberdeen AB25 2ZP, United Kingdom, TauRx Therapeutics Ltd., Singapore 068805, and the Department of Chemistry, University of Aberdeen, Aberdeen AB24 3UE, United Kingdom
| | - John M D Storey
- TauRx Therapeutics Ltd., Singapore 068805, and the Department of Chemistry, University of Aberdeen, Aberdeen AB24 3UE, United Kingdom
| | - Scott Clunas
- the Department of Chemistry, University of Aberdeen, Aberdeen AB24 3UE, United Kingdom
| | - Kathleen A Harrington
- From the School of Medicine and Dentistry, University of Aberdeen, Aberdeen AB25 2ZP, United Kingdom
| | - David Horsley
- From the School of Medicine and Dentistry, University of Aberdeen, Aberdeen AB25 2ZP, United Kingdom
| | - Ahtsham Ishaq
- the Department of Chemistry, University of Aberdeen, Aberdeen AB24 3UE, United Kingdom
| | - Steven J Kemp
- the Department of Chemistry, University of Aberdeen, Aberdeen AB24 3UE, United Kingdom
| | - Christopher P Larch
- the Department of Chemistry, University of Aberdeen, Aberdeen AB24 3UE, United Kingdom
| | - Colin Marshall
- the Department of Chemistry, University of Aberdeen, Aberdeen AB24 3UE, United Kingdom
| | - Sarah L Nicoll
- the Department of Chemistry, University of Aberdeen, Aberdeen AB24 3UE, United Kingdom
| | - Janet E Rickard
- From the School of Medicine and Dentistry, University of Aberdeen, Aberdeen AB25 2ZP, United Kingdom
| | - Michael Simpson
- the Department of Chemistry, University of Aberdeen, Aberdeen AB24 3UE, United Kingdom
| | - James P Sinclair
- the Department of Chemistry, University of Aberdeen, Aberdeen AB24 3UE, United Kingdom
| | - Lynda J Storey
- the Department of Chemistry, University of Aberdeen, Aberdeen AB24 3UE, United Kingdom
| | - Claude M Wischik
- From the School of Medicine and Dentistry, University of Aberdeen, Aberdeen AB25 2ZP, United Kingdom, TauRx Therapeutics Ltd., Singapore 068805, and
| |
Collapse
|
27
|
Benyair R, Ogen-Shtern N, Mazkereth N, Shai B, Ehrlich M, Lederkremer GZ. Mammalian ER mannosidase I resides in quality control vesicles, where it encounters its glycoprotein substrates. Mol Biol Cell 2014; 26:172-84. [PMID: 25411339 PMCID: PMC4294666 DOI: 10.1091/mbc.e14-06-1152] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
ER mannosidase I (ERManI) was found recently in the Golgi. This result is found to arise artificially from membrane disturbance in immunofluorescence methods. ERManI is located in novel vesicles to which substrates traffic and that converge at the ER-derived quality control compartment under ER stress. Endoplasmic reticulum α1,2 mannosidase I (ERManI), a central component of ER quality control and ER-associated degradation (ERAD), acts as a timer enzyme, modifying N-linked sugar chains of glycoproteins with time. This process halts glycoprotein folding attempts when necessary and targets terminally misfolded glycoproteins to ERAD. Despite the importance of ERManI in maintenance of glycoprotein quality control, fundamental questions regarding this enzyme remain controversial. One such question is the subcellular localization of ERManI, which has been suggested to localize to the ER membrane, the ER-derived quality control compartment (ERQC), and, surprisingly, recently to the Golgi apparatus. To try to clarify this controversy, we applied a series of approaches that indicate that ERManI is located, at the steady state, in quality control vesicles (QCVs) to which ERAD substrates are transported and in which they interact with the enzyme. Both endogenous and exogenously expressed ERManI migrate at an ER-like density on iodixanol gradients, suggesting that the QCVs are derived from the ER. The QCVs are highly mobile, displaying dynamics that are dependent on microtubules and COP-II but not on COP-I vesicle machinery. Under ER stress conditions, the QCVs converge in a juxtanuclear region, at the ERQC, as previously reported. Our results also suggest that ERManI is turned over by an active autophagic process. Of importance, we found that membrane disturbance, as is common in immunofluorescence methods, leads to an artificial appearance of ERManI in a Golgi pattern.
Collapse
Affiliation(s)
- Ron Benyair
- Department of Cell Research and Immunology, George Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel
| | - Navit Ogen-Shtern
- Department of Cell Research and Immunology, George Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel
| | - Niv Mazkereth
- Department of Cell Research and Immunology, George Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel
| | - Ben Shai
- Department of Cell Research and Immunology, George Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel
| | - Marcelo Ehrlich
- Department of Cell Research and Immunology, George Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel
| | - Gerardo Z Lederkremer
- Department of Cell Research and Immunology, George Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel
| |
Collapse
|
28
|
Mudhasani R, Kota KP, Retterer C, Tran JP, Whitehouse CA, Bavari S. High content image-based screening of a protease inhibitor library reveals compounds broadly active against Rift Valley fever virus and other highly pathogenic RNA viruses. PLoS Negl Trop Dis 2014; 8:e3095. [PMID: 25144302 PMCID: PMC4140764 DOI: 10.1371/journal.pntd.0003095] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2014] [Accepted: 07/03/2014] [Indexed: 12/20/2022] Open
Abstract
High content image-based screening was developed as an approach to test a protease inhibitor small molecule library for antiviral activity against Rift Valley fever virus (RVFV) and to determine their mechanism of action. RVFV is the causative agent of severe disease of humans and animals throughout Africa and the Arabian Peninsula. Of the 849 compounds screened, 34 compounds exhibited ≥50% inhibition against RVFV. All of the hit compounds could be classified into 4 distinct groups based on their unique chemical backbone. Some of the compounds also showed broad antiviral activity against several highly pathogenic RNA viruses including Ebola, Marburg, Venezuela equine encephalitis, and Lassa viruses. Four hit compounds (C795-0925, D011-2120, F694-1532 and G202-0362), which were most active against RVFV and showed broad-spectrum antiviral activity, were selected for further evaluation for their cytotoxicity, dose response profile, and mode of action using classical virological methods and high-content imaging analysis. Time-of-addition assays in RVFV infections suggested that D011-2120 and G202-0362 targeted virus egress, while C795-0925 and F694-1532 inhibited virus replication. We showed that D011-2120 exhibited its antiviral effects by blocking microtubule polymerization, thereby disrupting the Golgi complex and inhibiting viral trafficking to the plasma membrane during virus egress. While G202-0362 also affected virus egress, it appears to do so by a different mechanism, namely by blocking virus budding from the trans Golgi. F694-1532 inhibited viral replication, but also appeared to inhibit overall cellular gene expression. However, G202-0362 and C795-0925 did not alter any of the morphological features that we examined and thus may prove to be good candidates for antiviral drug development. Overall this work demonstrates that high-content image analysis can be used to screen chemical libraries for new antivirals and to determine their mechanism of action and any possible deleterious effects on host cellular biology. Rift Valley fever (RVF) is an arthropod-borne viral zoonosis that occurs in large parts of sub-Saharan and North Africa and in 2000 emerged outside the African continent for the first time, raising concerns that it could further expand its geographical range. The disease in humans can result in encephalitis or hemorrhagic fever and in ruminants often results in abortion in pregnant females. Due to the lack of a licensed and commercially available vaccine, efforts to discover effective antiviral drugs are underway. Drug discovery using high content image-based screening is an effective tool that has been successfully used to identify new drugs. In this study, we developed an image-based assay to identify compounds active against RVF virus and other highly pathogenic human viruses. We demonstrated the usefulness of our image-based high content assay in identifying potential RVF antivirals by screening a small subset of chemical compounds for inhibition of RVF virus in a human cell line (HeLa) and partially characterized their mechanism of action within infected cells. The methods we developed in this study will be useful in discovering new effective drugs to combat Rift Valley fever.
Collapse
Affiliation(s)
- Rajini Mudhasani
- Molecular and Translational Sciences Division, United States Army Medical Research Institute of Infectious Diseases, Frederick, Maryland, United States of America
| | - Krishna P. Kota
- Molecular and Translational Sciences Division, United States Army Medical Research Institute of Infectious Diseases, Frederick, Maryland, United States of America
| | - Cary Retterer
- Molecular and Translational Sciences Division, United States Army Medical Research Institute of Infectious Diseases, Frederick, Maryland, United States of America
| | - Julie P. Tran
- Molecular and Translational Sciences Division, United States Army Medical Research Institute of Infectious Diseases, Frederick, Maryland, United States of America
| | - Chris A. Whitehouse
- Molecular and Translational Sciences Division, United States Army Medical Research Institute of Infectious Diseases, Frederick, Maryland, United States of America
| | - Sina Bavari
- Molecular and Translational Sciences Division, United States Army Medical Research Institute of Infectious Diseases, Frederick, Maryland, United States of America
- * E-mail:
| |
Collapse
|
29
|
Yamamoto A. Gathering up meiotic telomeres: a novel function of the microtubule-organizing center. Cell Mol Life Sci 2014; 71:2119-34. [PMID: 24413667 PMCID: PMC11113538 DOI: 10.1007/s00018-013-1548-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2013] [Revised: 12/12/2013] [Accepted: 12/19/2013] [Indexed: 11/26/2022]
Abstract
During meiosis, telomeres cluster and promote homologous chromosome pairing. Telomere clustering depends on conserved SUN and KASH domain nuclear membrane proteins, which form a complex called the linker of nucleoskeleton and cytoskeleton (LINC) and connect telomeres with the cytoskeleton. It has been thought that LINC-mediated cytoskeletal forces induce telomere clustering. However, how cytoskeletal forces induce telomere clustering is not fully understood. Recent study of fission yeast has shown that the LINC complex forms the microtubule-organizing center (MTOC) at the telomere, which has been designated as the "telocentrosome", and that microtubule motors gather telomeres via telocentrosome-nucleated microtubules. This MTOC-dependent telomere clustering might be conserved in other eukaryotes. Furthermore, the MTOC-dependent clustering mechanism appears to function in various other biological events. This review presents an overview of the current understanding of the mechanism of meiotic telomere clustering and discusses the universality of the MTOC-dependent clustering mechanism.
Collapse
Affiliation(s)
- Ayumu Yamamoto
- Department of Chemistry, Graduate School of Science, Shizuoka University, 836 Ohya, Suruga-ku, Sizuoka, 422-8529, Japan,
| |
Collapse
|
30
|
Seixas E, Barros M, Seabra MC, Barral DC. Rab and Arf proteins in genetic diseases. Traffic 2013; 14:871-85. [PMID: 23565987 DOI: 10.1111/tra.12072] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2012] [Revised: 04/03/2013] [Accepted: 04/08/2013] [Indexed: 01/29/2023]
Abstract
Rab and ADP-ribosylation factor (Arf) family proteins are master regulators of membrane trafficking and are involved in all steps of vesicular transport. These families of small guanine-nucleotide-binding (G) proteins are well suited to regulate membrane trafficking processes since their nucleotide state determines their conformation and the capacity to bind to a multitude of effectors, which mediate their functions. In recent years, several inherited diseases have been associated with mutations in genes encoding proteins belonging to these two families or in proteins that regulate their GTP-binding cycle. The genetic diseases that are caused by defects in Rabs, Arfs or their regulatory proteins are heterogeneous and display diverse symptoms. However, these diseases mainly affect two types of subcellular compartments, namely lysosome-related organelles and cilia. Also, several of these diseases affect the nervous system. Thus, the study of these diseases represents an opportunity to understand their etiology and the molecular mechanisms involved, as well as to develop novel therapeutic strategies.
Collapse
Affiliation(s)
- Elsa Seixas
- CEDOC, Faculdade de Ciências Médicas, FCM, Universidade Nova de Lisboa, 1169-056, Lisboa, Portugal
| | | | | | | |
Collapse
|
31
|
Abstract
Small GTPases regulate a wide range of homeostatic processes such as cytoskeletal dynamics, organelle homeostasis, cell migration and vesicle trafficking, as well as in pathologic conditions such as carcinogenesis and metastatic spreading. Therefore, it is important to understand the regulation of small GTPase signaling, but this is complicated by the fact that crosstalk exists between different GTPase families and that we have to understand how they signal in time and space. The Golgi apparatus represents a hub for several signaling molecules and its importance in this field is constantly increasing. In this review we will discuss small GTPases signaling at the Golgi apparatus. Then, we will highlight recent work that contributed to a better understanding of crosstalk between different small GTPase families, with a special emphasis on their crosstalk at the Golgi apparatus. Finally, we will give a brief overview of available methods and tools to investigate spatio-temporal small GTPase crosstalk.
Collapse
Affiliation(s)
- Francesco Baschieri
- Department of Biology, University of Konstanz, Universitätsstrasse 10, D-78457 Konstanz, Germany
| | | |
Collapse
|
32
|
Cellular and Molecular Biology of Neuronal Dystonin. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2013; 300:85-120. [DOI: 10.1016/b978-0-12-405210-9.00003-5] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
|
33
|
Rab33a mediates anterograde vesicular transport for membrane exocytosis and axon outgrowth. J Neurosci 2012; 32:12712-25. [PMID: 22972995 DOI: 10.1523/jneurosci.0989-12.2012] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Axon outgrowth requires plasma membrane expansion, which results from post-Golgi vesicular transport and fusion. However, the molecular mechanisms regulating post-Golgi vesicular trafficking for membrane expansion and axon outgrowth remain unclear. Here, we show that Rab33a expression became upregulated during axon outgrowth of cultured rat hippocampal neurons. Rab33a was preferentially localized to the Golgi apparatus and to synaptophysin-positive vesicles that are transported along the growing axon. Previous studies showed that synaptophysin is localized to post-Golgi vesicles transported by fast axonal transport in developing neurons. Reduction of Rab33a expression by RNAi (RNA interference) inhibited the anterograde transport of synaptophysin-positive vesicles, leading to their decrease in axonal tips. Furthermore, this treatment reduced membrane fusion of synaptophysin-positive vesicles at the growth cones and inhibited axon outgrowth. Overexpression of Rab33a, on the other hand, induced excessive accumulation of synaptophysin-positive vesicles and concurrent formation of surplus axons. These data suggest that Rab33a participates in axon outgrowth by mediating anterograde axonal transport of synaptophysin-positive vesicles and their concomitant fusion at the growth cones.
Collapse
|
34
|
Abstract
Most organelles within the exocytic and endocytic pathways typically acidify their interiors, a phenomenon that is known to be crucial for their optimal functioning in eukaryotic cells. This review highlights recent advances in our understanding of how Golgi acidity is maintained and regulated, and how its misregulation contributes to organelle dysfunction and disease. Both its biosynthetic products (glycans) and protein-sorting events are highly sensitive to changes in Golgi luminal pH and are affected in certain human disease states such as cancers and cutis laxa. Other potential disease states that are caused by, or are associated with, Golgi pH misregulation will also be discussed.
Collapse
Affiliation(s)
- Antti Rivinoja
- Department of Biochemistry, University of Oulu, Oulu, Finland
| | | | | | | |
Collapse
|
35
|
Glotfelty LG, Hecht GA. Enteropathogenic E. coli effectors EspG1/G2 disrupt tight junctions: new roles and mechanisms. Ann N Y Acad Sci 2012; 1258:149-58. [PMID: 22731728 DOI: 10.1111/j.1749-6632.2012.06563.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Enteropathogenic E. coli (EPEC) infection is a major cause of infantile diarrhea in the developing world. Using a type-three secretion system, bacterial effector proteins are transferred to the host cell cytosol where they affect multiple physiological functions, ultimately leading to diarrheal disease. Disruption of intestinal epithelial cell tight junctions is a major consequence of EPEC infection and is mediated by multiple effector proteins, among them EspG1 and its homologue EspG2. EspG1/G2 contribute to loss of barrier function via an undefined mechanism that may be linked to their disruption of microtubule networks. Recently new investigations have identified additional roles for EspG. Sequestration of active ADP-ribosylating factor (ARF) proteins and promotion of p21-activated kinase (PAK) activity as well as inhibition of Golgi-mediated protein secretion have all been linked to EspG. In this review, we examine the functions of EspG1/G2 and discuss potential mechanisms of EspG-mediated tight junction disruption.
Collapse
Affiliation(s)
- Lila G Glotfelty
- Section of Digestive Diseases and Nutrition, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois, USA
| | | |
Collapse
|
36
|
Abstract
Cell polarization is an evolutionarily conserved process that facilitates asymmetric distribution of organelles and proteins and that is modified dynamically during physiological processes such as cell division, migration, and morphogenesis. The plasticity with which cells change their behavior and phenotype in response to cell intrinsic and extrinsic cues is an essential feature of normal physiology. In disease states such as cancer, cells lose their ability to behave normally in response to physiological cues. A molecular understanding of mechanisms that alter the behavior of cancer cells is limited. Cell polarity proteins are a recognized class of molecules that can receive and interpret both intrinsic and extrinsic signals to modulate cell behavior. In this review, we discuss how cell polarity proteins regulate a diverse array of biological processes and how they can contribute to alterations in the behavior of cancer cells.
Collapse
Affiliation(s)
- Senthil K Muthuswamy
- Ontario Cancer Institute, Campbell Family Institute for Breast Cancer Research, University of Toronto, Toronto M5G 2M9, Canada.
| | | |
Collapse
|
37
|
Millarte V, Farhan H. The Golgi in cell migration: regulation by signal transduction and its implications for cancer cell metastasis. ScientificWorldJournal 2012; 2012:498278. [PMID: 22623902 PMCID: PMC3353474 DOI: 10.1100/2012/498278] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2011] [Accepted: 12/18/2011] [Indexed: 01/17/2023] Open
Abstract
Migration and invasion are fundamental features of metastatic cancer cells. The Golgi apparatus, an organelle involved in posttranslational modification and sorting of proteins, is widely accepted to regulate directional cell migration. In addition, mounting evidence suggests that the Golgi is a hub for different signaling pathways. In this paper we will give an overview on how polarized secretion and microtubule nucleation at the Golgi regulate directional cell migration. We will review different signaling pathways that signal to and from the Golgi. Finally, we will discuss how these signaling pathways regulate the role of the Golgi in cell migration and invasion. We propose that by identifying regulators of the Golgi, we might be able to uncover unappreciated modulators of cell migration. Uncovering the regulatory network that orchestrates cell migration is of fundamental importance for the development of new therapeutic strategies against cancer cell metastasis.
Collapse
Affiliation(s)
- Valentina Millarte
- Department of Biology, University of Konstanz, Universitätsstrasse 10, Konstanz, Germany
| | | |
Collapse
|
38
|
Sugawara T, Nakatsu D, Kii H, Maiya N, Adachi A, Yamamoto A, Kano F, Murata M. PKCδ and ε regulate the morphological integrity of the ER–Golgi intermediate compartment (ERGIC) but not the anterograde and retrograde transports via the Golgi apparatus. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2012; 1823:861-75. [DOI: 10.1016/j.bbamcr.2012.01.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/29/2011] [Revised: 01/13/2012] [Accepted: 01/17/2012] [Indexed: 02/03/2023]
|
39
|
Martins GG, Kolega J. A role for microtubules in endothelial cell protrusion in three-dimensional matrices. Biol Cell 2012; 104:271-86. [PMID: 22211516 DOI: 10.1111/boc.201100088] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2011] [Accepted: 12/28/2011] [Indexed: 11/30/2022]
Abstract
BACKGROUND INFORMATION Most cells reside in vivo in a three-dimensional (3D) environment surrounded by extracellular matrix and other neighbouring cells, conditions that are different from those found by cells cultured in vitro on two-dimensional (2D) substrata. Cell morphology and behaviour are very different under these two different conditions, but the structural basis for these differences is still not understood, especially the role of microtubules (MTs). To address this issue, we studied the early spreading behaviour of bovine aortic endothelial cells (BAECs) cultured in 3D collagen matrices and on 2D substrata, in the presence of MT-disrupting drugs. RESULTS We found that depolymerisation of MTs greatly reduces the ability of BAECs to form large and stable protrusions inside 3D collagen matrices, an effect that is less pronounced when the cells are cultured on 2D substrata. Colcemid-treated BAECs inside 3D matrices begin assembling protrusions and pull on the matrix, but they fail to extend those protrusions deep into the matrix. It has been previously reported that MT disruption affects Rho signalling which may result in increased cell rigidity and adhesiveness to 2D matrices. Accordingly, we demonstrate that colcemid treatment indeed leads to activation of Rho-kinase (ROCK) targets, which in turn results in activation of regulatory myosin light chains, and that blocking of ROCK mitigates some of the effects of MT disruption in cell spreading in 3D. CONCLUSIONS Our results show that MT depolymerisation is particularly disruptive when cells interact with pliable 3D matrices, suggesting a role for MTs and the Rho pathway in the fine-tuning of contractile and adhesive forces necessary to sustain cell motility in vivo.
Collapse
Affiliation(s)
- Gabriel G Martins
- Centro de Biologia Ambiental/Departamento de Biologia Animal, Faculdade de Ciencias, Universidade de Lisboa, 1749-016 Lisbon, Portugal.
| | | |
Collapse
|
40
|
TRAPPC9 mediates the interaction between p150 and COPII vesicles at the target membrane. PLoS One 2012; 7:e29995. [PMID: 22279557 PMCID: PMC3261171 DOI: 10.1371/journal.pone.0029995] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2011] [Accepted: 12/07/2011] [Indexed: 11/19/2022] Open
Abstract
Background The transport of endoplasmic reticulum (ER)-derived COPII vesicles toward the ER-Golgi intermediate compartment (ERGIC) requires cytoplasmic dynein and is dependent on microtubules. p150Glued, a subunit of dynactin, has been implicated in the transport of COPII vesicles via its interaction with COPII coat components Sec23 and Sec24. However, whether and how COPII vesicle tether, TRAPP (Transport protein particle), plays a role in the interaction between COPII vesicles and microtubules is currently unknown. Principle Findings We address the functional relationship between COPII tether TRAPP and dynactin. Overexpressed TRAPP subunits interfered with microtubule architecture by competing p150Glued away from the MTOC. TRAPP subunit TRAPPC9 bound directly to p150Glued via the same carboxyl terminal domain of p150Glued that binds Sec23 and Sec24. TRAPPC9 also inhibited the interaction between p150Glued and Sec23/Sec24 both in vitro and in vivo, suggesting that TRAPPC9 serves to uncouple p150Glued from the COPII coat, and to relay the vesicle-dynactin interaction at the target membrane. Conclusions These findings provide a new perspective on the function of TRAPP as an adaptor between the ERGIC membrane and dynactin. By preserving the connection between dynactin and the tethered and/or fused vesicles, TRAPP allows nascent ERGIC to continue the movement along the microtubules as they mature into the cis-Golgi.
Collapse
|
41
|
Golgi apparatus fragmentation as a mechanism responsible for uniform delivery of uroplakins to the apical plasma membrane of uroepithelial cells. Biol Cell 2012; 102:593-607. [DOI: 10.1042/bc20100024] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
42
|
Zaal KJM, Reid E, Mousavi K, Zhang T, Mehta A, Bugnard E, Sartorelli V, Ralston E. Who needs microtubules? Myogenic reorganization of MTOC, Golgi complex and ER exit sites persists despite lack of normal microtubule tracks. PLoS One 2011; 6:e29057. [PMID: 22216166 PMCID: PMC3246457 DOI: 10.1371/journal.pone.0029057] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2011] [Accepted: 11/20/2011] [Indexed: 11/18/2022] Open
Abstract
A wave of structural reorganization involving centrosomes, microtubules, Golgi complex and ER exit sites takes place early during skeletal muscle differentiation and completely remodels the secretory pathway. The mechanism of these changes and their functional implications are still poorly understood, in large part because all changes occur seemingly simultaneously. In an effort to uncouple the reorganizations, we have used taxol, nocodazole, and the specific GSK3-β inhibitor DW12, to disrupt the dynamic microtubule network of differentiating cultures of the mouse skeletal muscle cell line C2. Despite strong effects on microtubules, cell shape and cell fusion, none of the treatments prevented early differentiation. Redistribution of centrosomal proteins, conditional on differentiation, was in fact increased by taxol and nocodazole and normal in DW12. Redistributions of Golgi complex and ER exit sites were incomplete but remained tightly linked under all circumstances, and conditional on centrosomal reorganization. We were therefore able to uncouple microtubule reorganization from the other events and to determine that centrosomal proteins lead the reorganization hierarchy. In addition, we have gained new insight into structural and functional aspects of the reorganization of microtubule nucleation during myogenesis.
Collapse
Affiliation(s)
- Kristien J M Zaal
- Light Imaging Section, Office of Science and Technology, National Institute of Arthritis, Musculoskeletal, and Skin Disease, National Institutes of Health, Bethesda, Maryland, USA.
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Kasap M, Karaoz E, Akpinar G, Aksoy A, Erman G. A unique Golgi apparatus distribution may be a marker for osteogenic differentiation of hDP-MSCs. Cell Biochem Funct 2011; 29:489-95. [DOI: 10.1002/cbf.1776] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2011] [Revised: 05/20/2011] [Accepted: 06/03/2011] [Indexed: 01/07/2023]
Affiliation(s)
- Murat Kasap
- Department of Medical Biology/KABI Proteomics Laboratory; Kocaeli University Medical School; Kocaeli; Turkey
| | - Erdal Karaoz
- Applied Stem Cell and Gene Therapy Research Center; Kocaeli University; Kocaeli; Turkey
| | - Gurler Akpinar
- Department of Medical Biology/KABI Proteomics Laboratory; Kocaeli University Medical School; Kocaeli; Turkey
| | - Ayca Aksoy
- Applied Stem Cell and Gene Therapy Research Center; Kocaeli University; Kocaeli; Turkey
| | - Gulay Erman
- Applied Stem Cell and Gene Therapy Research Center; Kocaeli University; Kocaeli; Turkey
| |
Collapse
|
44
|
Yessotoxin as an apoptotic inducer. Toxicon 2011; 57:947-58. [DOI: 10.1016/j.toxicon.2011.03.012] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2010] [Revised: 01/31/2011] [Accepted: 03/14/2011] [Indexed: 12/12/2022]
|
45
|
Lee S, Sunil N, Tejada JM, Shea TB. Differential roles of kinesin and dynein in translocation of neurofilaments into axonal neurites. J Cell Sci 2011; 124:1022-31. [PMID: 21363889 DOI: 10.1242/jcs.079046] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Neurofilament (NF) subunits translocate within axons as short NFs, non-filamentous punctate structures ('puncta') and diffuse material that might comprise individual subunits and/or oligomers. Transport of NFs into and along axons is mediated by the microtubule (MT) motor proteins kinesin and dynein. Despite being characterized as a retrograde motor, dynein nevertheless participates in anterograde NF transport through associating with long MTs or the actin cortex through its cargo domain; relatively shorter MTs associated with the motor domain are then propelled in an anterograde direction, along with any linked NFs. Here, we show that inhibition of dynein function, through dynamitin overexpression or intracellular delivery of anti-dynein antibody, selectively reduced delivery of GFP-tagged short NFs into the axonal hillock, with a corresponding increase in the delivery of puncta, suggesting that dynein selectively delivered short NFs into axonal neurites. Nocodazole-mediated depletion of short MTs had the same effect. By contrast, intracellular delivery of anti-kinesin antibody inhibited anterograde transport of short NFs and puncta to an equal extent. These findings suggest that anterograde axonal transport of linear NFs is more dependent upon association with translocating MTs (which are themselves translocated by dynein) than is transport of NF puncta or oligomers.
Collapse
Affiliation(s)
- Sangmook Lee
- Center for Cellular Neurobiology and Neurodegeneration Research, Department of Biological Sciences, University of Massachusetts, One University Avenue, Lowell, MA 01854, USA
| | | | | | | |
Collapse
|
46
|
Cameron RS, Arvan P, Castle JD. Secretory Membranes and the Exocrine Storage Compartment. Compr Physiol 2011. [DOI: 10.1002/cphy.cp060307] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
47
|
Dippold HC, Ng MM, Farber-Katz SE, Lee SK, Kerr ML, Peterman MC, Sim R, Wiharto PA, Galbraith KA, Madhavarapu S, Fuchs GJ, Meerloo T, Farquhar MG, Zhou H, Field SJ. GOLPH3 bridges phosphatidylinositol-4- phosphate and actomyosin to stretch and shape the Golgi to promote budding. Cell 2009; 139:337-51. [PMID: 19837035 DOI: 10.1016/j.cell.2009.07.052] [Citation(s) in RCA: 491] [Impact Index Per Article: 30.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2008] [Revised: 04/13/2009] [Accepted: 07/31/2009] [Indexed: 11/15/2022]
Abstract
Golgi membranes, from yeast to humans, are uniquely enriched in phosphatidylinositol-4-phosphate (PtdIns(4)P), although the role of this lipid remains poorly understood. Using a proteomic lipid-binding screen, we identify the Golgi protein GOLPH3 (also called GPP34, GMx33, MIDAS, or yeast Vps74p) as a PtdIns(4)P-binding protein that depends on PtdIns(4)P for its Golgi localization. We further show that GOLPH3 binds the unconventional myosin MYO18A, thus connecting the Golgi to F-actin. We demonstrate that this linkage is necessary for normal Golgi trafficking and morphology. The evidence suggests that GOLPH3 binds to PtdIns(4)P-rich trans-Golgi membranes and MYO18A conveying a tensile force required for efficient tubule and vesicle formation. Consequently, this tensile force stretches the Golgi into the extended ribbon observed by fluorescence microscopy and the familiar flattened form observed by electron microscopy.
Collapse
Affiliation(s)
- Holly C Dippold
- Division of Endocrinology and Metabolism, Department of Medicine, University of California, San Diego, La Jolla, CA 92093-0707, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Blain EJ. Involvement of the cytoskeletal elements in articular cartilage homeostasis and pathology. Int J Exp Pathol 2009; 90:1-15. [PMID: 19200246 DOI: 10.1111/j.1365-2613.2008.00625.x] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
The cytoskeleton of all cells is a three-dimensional network comprising actin microfilaments, tubulin microtubules and intermediate filaments. Studies in many cell types have indicated roles for these cytoskeletal proteins in many diverse cellular processes including alteration of cell shape, movement of organelles, migration, endocytosis, secretion, cell division and extracellular matrix assembly. The cytoskeletal networks are highly organized in structure enabling them to fulfil their biological functions. This review will primarily focus on the organization and function of the three major cytoskeletal networks in articular cartilage chondrocytes. Articular cartilage is a major load-bearing tissue of the synovial joint; it is well known that the cytoskeleton acts as a physical interface between the chondrocytes and the extracellular matrix in 'sensing' mechanical stimuli. The effect of mechanical load on cytoskeletal element expression and organization will also be reviewed. Abnormal mechanical load is widely believed to be a risk factor for the development of osteoarthritis. Several studies have intimated that the major cytoskeletal networks are disorganized or often absent in osteoarthritic cartilage chondrocytes. The implications and possible reasoning for this are more widely discussed and placed into context with their potential relevance to disease and therapeutic strategies.
Collapse
Affiliation(s)
- Emma J Blain
- Connective Tissue Biology Laboratories, School of Biosciences, Cardiff University, Cardiff, UK.
| |
Collapse
|
49
|
Nesprin 4 is an outer nuclear membrane protein that can induce kinesin-mediated cell polarization. Proc Natl Acad Sci U S A 2009; 106:2194-9. [PMID: 19164528 DOI: 10.1073/pnas.0808602106] [Citation(s) in RCA: 272] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Nucleocytoplasmic coupling is mediated by outer nuclear membrane (ONM) nesprin proteins and inner nuclear membrane Sun proteins. Interactions spanning the perinuclear space create nesprin-Sun complexes connecting the cytoskeleton to nuclear components. A search for proteins displaying a conserved C-terminal sequence present in nesprins 1-3 identified nesprin 4 (Nesp4), a new member of this family. Nesp4 is a kinesin-1-binding protein that displays Sun-dependent localization to the ONM. Expression of Nesp4 is associated with dramatic changes in cellular organization involving relocation of the centrosome and Golgi apparatus relative to the nucleus. These effects can be accounted for entirely by Nesp4's kinesin-binding function. The implication is that Nesp4 may contribute to microtubule-dependent nuclear positioning.
Collapse
|
50
|
Bertrand L, Pearson A. The conserved N-terminal domain of herpes simplex virus 1 UL24 protein is sufficient to induce the spatial redistribution of nucleolin. J Gen Virol 2008; 89:1142-1151. [PMID: 18420791 DOI: 10.1099/vir.0.83573-0] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
UL24 is widely conserved among herpesviruses but its function during infection is poorly understood. Previously, we discovered a genetic link between UL24 and the herpes simplex virus 1-induced dispersal of the nucleolar protein nucleolin. Here, we report that in the absence of viral infection, transiently expressed UL24 accumulated in both the nucleus and the Golgi apparatus. In the majority of transfected cells, nuclear staining for UL24 was diffuse, but a minor staining pattern, whereby UL24 was present in nuclear foci corresponding to nucleoli, was also observed. Expression of UL24 correlated with the dispersal of nucleolin. This dispersal did not appear to be a consequence of a general disaggregation of nucleoli, as foci of fibrillarin staining persisted in cells expressing UL24. The conserved N-terminal region of UL24 was sufficient to cause this change in subcellular distribution of nucleolin. Interestingly, a bipartite nuclear localization signal predicted within the C terminus of UL24 was dispensable for nuclear localization. None of the five individual UL24 homology domains was required for nuclear or Golgi localization, but deletion of these domains resulted in the loss of nucleolin-dispersal activity. We determined that a nucleolar-targeting signal was contained within the first 60 aa of UL24. Our results show that the conserved N-terminal domain of UL24 is sufficient to specifically induce dispersal of nucleolin in the absence of other viral proteins or virus-induced cellular modifications. These results suggest that UL24 directly targets cellular factors that affect the composition of nucleoli.
Collapse
Affiliation(s)
- Luc Bertrand
- INRS-Institut Armand-Frappier, Université du Québec, Laval, QC H7V 1B7, Canada
| | - Angela Pearson
- INRS-Institut Armand-Frappier, Université du Québec, Laval, QC H7V 1B7, Canada
| |
Collapse
|