1
|
Scholey JM. Mitotic spindle membranes. Mol Biol Cell 2025; 36:re1. [PMID: 40067152 PMCID: PMC12005112 DOI: 10.1091/mbc.e24-10-0475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 01/31/2025] [Accepted: 02/05/2025] [Indexed: 04/19/2025] Open
Abstract
The mitotic spindle, which uses microtubules (MTs) and MT-based motor proteins to separate sister chromosomes prior to cell division, contains abundant membranes, organelles, and protein assemblies derived from the familiar interphase intracellular membrane network. In this essay, mainly with reference to selected animal and fungal cells, I summarize current ideas about the reciprocal functional relationship between these mitotic spindle-associated membranes and the spindle MT cytoskeleton, in which; 1) spindle membranes control the composition, Ca++ ion concentration and mechanical performance of the spindle MT cytoskeleton; and conversely 2) the spindle MT cytoskeleton contributes to membrane/organelle partitioning and inheritance during cell division and serves as a reservoir of membranes, organelles, and vesicles for delivery to the interphase cytoplasm, plasma membrane, and cleavage furrow.
Collapse
Affiliation(s)
- Jonathan M. Scholey
- Department of Molecular and Cell Biology, University of California at Davis, Davis, CA 95616
| |
Collapse
|
2
|
Zhang Z, Chen J, Ma R, Xu C, Lu Y, Zhou J, Xia K, Lu P. Tight Junction Component Occludin Binds to FIP5 to Regulate Endosome Trafficking and Mitotic Spindle Function. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2308822. [PMID: 38884279 PMCID: PMC11321699 DOI: 10.1002/advs.202308822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 04/16/2024] [Indexed: 06/18/2024]
Abstract
The genetic basis of vertebrate emergence during metazoan evolution has remained largely unknown. Understanding vertebrate-specific genes, such as the tight junction protein Occludin (Ocln), may help answer this question. Here, it is shown that mammary glands lacking Ocln exhibit retarded epithelial branching, owing to reduced cell proliferation and surface expansion. Interestingly, Ocln regulates mitotic spindle orientation and function, and its loss leads to a range of defects, including prolonged prophase and failed nuclear and/or cytoplasmic division. Mechanistically, Ocln binds to the RabGTPase-11 adaptor FIP5 and recruits recycling endosomes to the centrosome to participate in spindle assembly and function. FIP5 loss recapitulates Ocln null, leading to prolonged prophase, reduced cell proliferation, and retarded epithelial branching. These results identify a novel role in OCLN-mediated endosomal trafficking and potentially highlight its involvement in mediating membranous vesicle trafficking and function, which is evolutionarily conserved and essential.
Collapse
Affiliation(s)
- Zichao Zhang
- MOE Key Lab of Rare Pediatric DiseasesHengyang Medical SchoolUniversity of South ChinaHengyangChina
- Institute of Cytology and GeneticsSchool of Basic Medical SciencesHengyang Medical SchoolUniversity of South ChinaHengyangChina
- Institute for Future SciencesHengyang Medical SchoolUniversity of South ChinaChangshaChina
| | - Jing Chen
- MOE Key Lab of Rare Pediatric DiseasesHengyang Medical SchoolUniversity of South ChinaHengyangChina
- Institute of Cytology and GeneticsSchool of Basic Medical SciencesHengyang Medical SchoolUniversity of South ChinaHengyangChina
- Institute for Future SciencesHengyang Medical SchoolUniversity of South ChinaChangshaChina
| | - Rongze Ma
- MOE Key Lab of Rare Pediatric DiseasesHengyang Medical SchoolUniversity of South ChinaHengyangChina
- Institute of Cytology and GeneticsSchool of Basic Medical SciencesHengyang Medical SchoolUniversity of South ChinaHengyangChina
- Institute for Future SciencesHengyang Medical SchoolUniversity of South ChinaChangshaChina
| | - Chongshen Xu
- MOE Key Lab of Rare Pediatric DiseasesHengyang Medical SchoolUniversity of South ChinaHengyangChina
- Institute of Cytology and GeneticsSchool of Basic Medical SciencesHengyang Medical SchoolUniversity of South ChinaHengyangChina
- Institute for Future SciencesHengyang Medical SchoolUniversity of South ChinaChangshaChina
| | - Yunzhe Lu
- School of Life Science and TechnologyShanghaiTech UniversityShanghaiChina
| | - Jiecan Zhou
- The First Affiliated HospitalHengyang Medical SchoolUniversity of South ChinaHengyangChina
| | - Kun Xia
- MOE Key Lab of Rare Pediatric DiseasesHengyang Medical SchoolUniversity of South ChinaHengyangChina
- Institute of Cytology and GeneticsSchool of Basic Medical SciencesHengyang Medical SchoolUniversity of South ChinaHengyangChina
| | - Pengfei Lu
- MOE Key Lab of Rare Pediatric DiseasesHengyang Medical SchoolUniversity of South ChinaHengyangChina
- Institute of Cytology and GeneticsSchool of Basic Medical SciencesHengyang Medical SchoolUniversity of South ChinaHengyangChina
- Institute for Future SciencesHengyang Medical SchoolUniversity of South ChinaChangshaChina
| |
Collapse
|
3
|
Lee J, Miyagishima SY, Bhattacharya D, Yoon HS. From dusk till dawn: cell cycle progression in the red seaweed Gracilariopsis chorda (Rhodophyta). iScience 2024; 27:110190. [PMID: 38984202 PMCID: PMC11231608 DOI: 10.1016/j.isci.2024.110190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 04/29/2024] [Accepted: 06/03/2024] [Indexed: 07/11/2024] Open
Abstract
The conserved eukaryotic functions of cell cycle genes have primarily been studied using animal/plant models and unicellular algae. Cell cycle progression and its regulatory components in red (Rhodophyta) seaweeds are poorly understood. We analyzed diurnal gene expression data to investigate the cell cycle in the red seaweed Gracilariopsis chorda. We identified cell cycle progression and transitions in G. chorda which are induced by interactions of key regulators such as E2F/DP, RBR, cyclin-dependent kinases, and cyclins from dusk to dawn. However, several typical CDK inhibitor proteins are absent in red seaweeds. Interestingly, the G1-S transition in G. chorda is controlled by delayed transcription of GINS subunit 3. We propose that the delayed S phase entry in this seaweed may have evolved to minimize DNA damage (e.g., due to UV radiation) during replication. Our results provide important insights into cell cycle-associated physiology and its molecular mechanisms in red seaweeds.
Collapse
Affiliation(s)
- JunMo Lee
- Department of Oceanography, Kyungpook National University, Daegu 41566, Korea
- Kyungpook Institute of Oceanography, Kyungpook National University, Daegu 41566, Korea
| | - Shin-ya Miyagishima
- Department of Gene Function and Phenomics, National Institute of Genetics, Mishima, Shizuoka 411-8540, Japan
- Department of Genetics, Graduate University for Advanced Studies, SOKENDAI, Mishima, Shizuoka 411-8540, Japan
| | - Debashish Bhattacharya
- Department of Biochemistry and Microbiology, Rutgers University, New Brunswick, NJ 08901, USA
| | - Hwan Su Yoon
- Department of Biological Sciences, Sungkyunkwan University, Suwon 16419, Korea
| |
Collapse
|
4
|
Pandey A, Roy JK. Rab11 maintains the undifferentiated state of adult midgut precursors via DPP pathway. Exp Cell Res 2024; 439:114092. [PMID: 38754617 DOI: 10.1016/j.yexcr.2024.114092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 05/10/2024] [Accepted: 05/12/2024] [Indexed: 05/18/2024]
Abstract
Asymmetric stem cell divisions play instrumental roles in the maintenance, growth and differentiation of organs. Failure of asymmetric stem cell divisions may result in an array of developmental disorders, including cancer. It is well established that the gene, inscuteable, acts as the upstream component of asymmetric cell divisions. In Drosophila larval midgut, a founder adult midgut precursor (AMP) experiences an asymmetric division to instruct its first daughter to become a peripheral cell that serves as a niche where the AMP and its future daughters can remain undifferentiated. The present study demonstrates that inscuteable expressing stem cells require Rab11, a conserved small Ras-like GTPase, for proper proliferation and differentiation. As insc-GAL4 mediated Rab11RNAi in Drosophila larval and adult midguts show the disruption of the niche microenvironment of adult midgut precursors as well as elevated DPP signalling at the larval stage, which is associated with aberrant over-proliferation and early differentiation of larval AMPs and adult intestinal stem cells. The observed connections between Rab11, larval AMP proliferation, niche establishment, and DPP signalling highlight the potential for Rab11 to serve as a key regulatory factor in maintaining tissue homeostasis and balanced cellular growth.
Collapse
Affiliation(s)
- Akanksha Pandey
- Cytogenetics Laboratory, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, 221005, India
| | - Jagat Kumar Roy
- Cytogenetics Laboratory, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, 221005, India.
| |
Collapse
|
5
|
Rollins KR, Blankenship JT. Dysregulation of the endoplasmic reticulum blocks recruitment of centrosome-associated proteins resulting in mitotic failure. Development 2023; 150:dev201917. [PMID: 37971218 PMCID: PMC10690056 DOI: 10.1242/dev.201917] [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: 04/24/2023] [Accepted: 10/17/2023] [Indexed: 11/19/2023]
Abstract
The endoplasmic reticulum (ER) undergoes a remarkable transition in morphology during cell division to aid in the proper portioning of the ER. However, whether changes in ER behaviors modulate mitotic events is less clear. Like many animal embryos, the early Drosophila embryo undergoes rapid cleavage cycles in a lipid-rich environment. Here, we show that mitotic spindle formation, centrosomal maturation, and ER condensation occur with similar time frames in the early syncytium. In a screen for Rab family GTPases that display dynamic function at these stages, we identified Rab1. Rab1 disruption led to an enhanced buildup of ER at the spindle poles and produced an intriguing 'mini-spindle' phenotype. ER accumulation around the mitotic space negatively correlates with spindle length/intensity. Importantly, centrosomal maturation is defective in these embryos, as mitotic recruitment of key centrosomal proteins is weakened after Rab1 disruption. Finally, division failures and ER overaccumulation is rescued by Dynein inhibition, demonstrating that Dynein is essential for ER spindle recruitment. These results reveal that ER levels must be carefully tuned during mitotic processes to ensure proper assembly of the division machinery.
Collapse
Affiliation(s)
| | - J. Todd Blankenship
- Department of Biological Sciences, University of Denver, Denver, CO 80208, USA
| |
Collapse
|
6
|
Guo RJ, Cao YF, Li EM, Xu LY. Multiple functions and dual characteristics of RAB11A in cancers. Biochim Biophys Acta Rev Cancer 2023; 1878:188966. [PMID: 37657681 DOI: 10.1016/j.bbcan.2023.188966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 08/05/2023] [Accepted: 08/05/2023] [Indexed: 09/03/2023]
Abstract
Vesicle trafficking is an unceasing and elaborate cellular process that functions in material transport and information delivery. Recent studies have identified the small GTPase, Ras-related protein in brain 11A (RAB11A), as a key regulator in this process. Aberrant RAB11A expression has been reported in several types of cancers, suggesting the important functions and characteristics of RAB11A in cancer. These discoveries are of great significance because therapeutic strategies based on the physiological and pathological status of RAB11A might make cancer treatment more effective, as the molecular mechanisms of cancer development have not been completely revealed. However, these studies on RAB11A have not been reviewed and discussed specifically. Therefore, we summarize and discuss the recent findings of RAB11A involvement in different biological processes, including endocytic recycling regulation, receptors and adhesion molecules recycling, exosome secretion, phagophore formation and cytokinesis, as well as regulatory mechanisms in several tumor types. Moreover, contradictory effects of RAB11A have also been observed in different types of cancers, implying the dual characteristics of RAB11A in cancer, which are either oncogenic or tumor-suppressive. This review on the functions and characteristics of RAB11A highlights the value of RAB11A in inducing multiple important phenotypes based on vesicle trafficking and therefore will offer insights for future studies to reveal the molecular mechanisms, clinical significance, and therapeutic targeting of RAB11A in different cancers.
Collapse
Affiliation(s)
- Rui-Jian Guo
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou 515041, Guangdong, PR China
| | - Yu-Fei Cao
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou 515041, Guangdong, PR China
| | - En-Min Li
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou 515041, Guangdong, PR China.
| | - Li-Yan Xu
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou 515041, Guangdong, PR China; Institute of Oncologic Pathology, Shantou University Medical College, Shantou 515041, Guangdong, PR China.
| |
Collapse
|
7
|
Joseph I, Flores J, Farrell V, Davis J, Bianchi‐Smak J, Feng Q, Goswami S, Lin X, Wei Z, Tong K, Feng Z, Verzi MP, Bonder EM, Goldenring JR, Gao N. RAB11A and RAB11B control mitotic spindle function in intestinal epithelial progenitor cells. EMBO Rep 2023; 24:e56240. [PMID: 37424454 PMCID: PMC10481667 DOI: 10.15252/embr.202256240] [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: 10/05/2022] [Revised: 06/20/2023] [Accepted: 06/27/2023] [Indexed: 07/11/2023] Open
Abstract
RAB11 small GTPases and associated recycling endosome have been localized to mitotic spindles and implicated in regulating mitosis. However, the physiological significance of such regulation has not been observed in mammalian tissues. We have used newly engineered mouse models to investigate intestinal epithelial renewal in the absence of single or double isoforms of RAB11 family members: Rab11a and Rab11b. Comparing with single knockouts, mice with compound ablation demonstrate a defective cell cycle entry and robust mitotic arrest followed by apoptosis, leading to a total penetrance of lethality within 3 days of gene ablation. Upon Rab11 deletion ex vivo, enteroids show abnormal mitotic spindle formation and cell death. Untargeted proteomic profiling of Rab11a and Rab11b immunoprecipitates has uncovered a shared interactome containing mitotic spindle microtubule regulators. Disrupting Rab11 alters kinesin motor KIF11 function and impairs bipolar spindle formation and cell division. These data demonstrate that RAB11A and RAB11B redundantly control mitotic spindle function and intestinal progenitor cell division, a mechanism that may be utilized to govern the homeostasis and renewal of other mammalian tissues.
Collapse
Affiliation(s)
- Ivor Joseph
- Department of Biological SciencesRutgers UniversityNewarkNJUSA
| | - Juan Flores
- Department of Biological SciencesRutgers UniversityNewarkNJUSA
| | | | - Justin Davis
- Department of Biological SciencesRutgers UniversityNewarkNJUSA
| | | | - Qiang Feng
- Department of Biological SciencesRutgers UniversityNewarkNJUSA
| | | | - Xiang Lin
- Department of Computer SciencesNew Jersey Institute of TechnologyNewarkNJUSA
| | - Zhi Wei
- Department of Computer SciencesNew Jersey Institute of TechnologyNewarkNJUSA
| | - Kevin Tong
- Department of GeneticsRutgers UniversityNew BrunswickNJUSA
| | - Zhaohui Feng
- Rutgers Cancer Institute of New JerseyNew BrunswickNJUSA
| | | | - Edward M Bonder
- Department of Biological SciencesRutgers UniversityNewarkNJUSA
| | - James R Goldenring
- Section of Surgical Sciences and Epithelial Biology CenterVanderbilt University Medical CenterNashvilleTNUSA
| | - Nan Gao
- Department of Biological SciencesRutgers UniversityNewarkNJUSA
| |
Collapse
|
8
|
Malycheva D, Alvarado-Kristensson M. Centrosome Movements Are TUBG1-Dependent. Int J Mol Sci 2023; 24:13154. [PMID: 37685969 PMCID: PMC10488117 DOI: 10.3390/ijms241713154] [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: 07/22/2023] [Revised: 08/16/2023] [Accepted: 08/22/2023] [Indexed: 09/10/2023] Open
Abstract
The centrosome of mammalian cells is in constant movement and its motion plays a part in cell differentiation and cell division. The purpose of this study was to establish the involvement of the TUBG meshwork in centrosomal motility. In live cells, we used a monomeric red-fluorescence-protein-tagged centrin 2 gene and a green-fluorescence-protein-tagged TUBG1 gene for labeling the centrosome and the TUBG1 meshwork, respectively. We found that centrosome movements occurred in cellular sites rich in GTPase TUBG1 and single-guide RNA mediated a reduction in the expression of TUBG1, altering the motility pattern of centrosomes. We propose that the TUBG1 meshwork enables the centrosomes to move by providing them with an interacting platform that mediates positional changes. These findings uncover a novel regulatory mechanism that controls the behavior of centrosomes.
Collapse
Affiliation(s)
| | - Maria Alvarado-Kristensson
- Molecular Pathology, Department of Translational Medicine, Skåne University Hospital, Lund University, 21428 Malmö, Sweden;
| |
Collapse
|
9
|
Crellin HA, Buckley CE. Using Optogenetics to Investigate the Shared Mechanisms of Apical-Basal Polarity and Mitosis. Cells Tissues Organs 2023; 213:161-180. [PMID: 36599311 DOI: 10.1159/000528796] [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: 07/25/2022] [Accepted: 12/18/2022] [Indexed: 01/05/2023] Open
Abstract
The initiation of apical-basal (AB) polarity and the process of mitotic cell division are both characterised by the generation of specialised plasma membrane and cortical domains. These are generated using shared mechanisms, such as asymmetric protein accumulation, Rho GTPase signalling, cytoskeletal reorganisation, vesicle trafficking, and asymmetric phosphoinositide distribution. In epithelial tissue, the coordination of AB polarity and mitosis in space and time is important both during initial epithelial development and to maintain tissue integrity and ensure appropriate cell differentiation at later stages. Whilst significant progress has been made in understanding the mechanisms underlying cell division and AB polarity, it has so far been challenging to fully unpick the complex interrelationship between polarity, signalling, morphogenesis, and cell division. However, the recent emergence of optogenetic protein localisation techniques is now allowing researchers to reversibly control protein activation, localisation, and signalling with high spatiotemporal resolution. This has the potential to revolutionise our understanding of how subcellular processes such as AB polarity are integrated with cell behaviours such as mitosis and how these processes impact whole tissue morphogenesis. So far, these techniques have been used to investigate processes such as cleavage furrow ingression, mitotic spindle positioning, and in vivo epithelial morphogenesis. This review describes some of the key shared mechanisms of cell division and AB polarity establishment, how they are coordinated during development and how the advance of optogenetic techniques is furthering this research field.
Collapse
Affiliation(s)
- Helena A Crellin
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Clare E Buckley
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| |
Collapse
|
10
|
Krishnan N, Swoger M, Rathbun LI, Fioramonti PJ, Freshour J, Bates M, Patteson AE, Hehnly H. Rab11 endosomes and Pericentrin coordinate centrosome movement during pre-abscission in vivo. Life Sci Alliance 2022; 5:e202201362. [PMID: 35304423 PMCID: PMC8933627 DOI: 10.26508/lsa.202201362] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 03/02/2022] [Accepted: 03/03/2022] [Indexed: 11/24/2022] Open
Abstract
The last stage of cell division involves two daughter cells remaining interconnected by a cytokinetic bridge that is cleaved during abscission. Conserved between the zebrafish embryo and human cells, we found that the oldest centrosome moves in a Rab11-dependent manner towards the cytokinetic bridge sometimes followed by the youngest. Rab11-endosomes are organized in a Rab11-GTP dependent manner at the mother centriole during pre-abscission, with Rab11 endosomes at the oldest centrosome being more mobile compared with the youngest. The GTPase activity of Rab11 is necessary for the centrosome protein, Pericentrin, to be enriched at the centrosome. Reduction in Pericentrin expression or optogenetic disruption of Rab11-endosome function inhibited both centrosome movement towards the cytokinetic bridge and abscission, resulting in daughter cells prone to being binucleated and/or having supernumerary centrosomes. These studies suggest that Rab11-endosomes contribute to centrosome function during pre-abscission by regulating Pericentrin organization resulting in appropriate centrosome movement towards the cytokinetic bridge and subsequent abscission.
Collapse
Affiliation(s)
- Nikhila Krishnan
- Department of Biology, Syracuse University, Syracuse, NY, USA
- BioInspired Institute, Syracuse University, Syracuse, NY, USA
| | - Maxx Swoger
- Department of Physics, Syracuse University, Physics Building, Syracuse, NY, USA
- BioInspired Institute, Syracuse University, Syracuse, NY, USA
| | - Lindsay I Rathbun
- Department of Biology, Syracuse University, Syracuse, NY, USA
- BioInspired Institute, Syracuse University, Syracuse, NY, USA
| | - Peter J Fioramonti
- Department of Biology, Syracuse University, Syracuse, NY, USA
- BioInspired Institute, Syracuse University, Syracuse, NY, USA
| | - Judy Freshour
- Department of Biology, Syracuse University, Syracuse, NY, USA
- BioInspired Institute, Syracuse University, Syracuse, NY, USA
| | - Michael Bates
- Department of Biology, Syracuse University, Syracuse, NY, USA
- BioInspired Institute, Syracuse University, Syracuse, NY, USA
| | - Alison E Patteson
- Department of Physics, Syracuse University, Physics Building, Syracuse, NY, USA
- BioInspired Institute, Syracuse University, Syracuse, NY, USA
| | - Heidi Hehnly
- Department of Biology, Syracuse University, Syracuse, NY, USA
- BioInspired Institute, Syracuse University, Syracuse, NY, USA
| |
Collapse
|
11
|
Fine-tuning cell organelle dynamics during mitosis by small GTPases. Front Med 2022; 16:339-357. [PMID: 35759087 DOI: 10.1007/s11684-022-0926-1] [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: 10/26/2021] [Accepted: 02/24/2022] [Indexed: 11/04/2022]
Abstract
During mitosis, the allocation of genetic material concurs with organelle transformation and distribution. The coordination of genetic material inheritance with organelle dynamics directs accurate mitotic progression, cell fate determination, and organismal homeostasis. Small GTPases belonging to the Ras superfamily regulate various cell organelles during division. Being the key regulators of membrane dynamics, the dysregulation of small GTPases is widely associated with cell organelle disruption in neoplastic and non-neoplastic diseases, such as cancer and Alzheimer's disease. Recent discoveries shed light on the molecular properties of small GTPases as sophisticated modulators of a remarkably complex and perfect adaptors for rapid structure reformation. This review collects current knowledge on small GTPases in the regulation of cell organelles during mitosis and highlights the mediator role of small GTPase in transducing cell cycle signaling to organelle dynamics during mitosis.
Collapse
|
12
|
Akhmanova A, Kapitein LC. Mechanisms of microtubule organization in differentiated animal cells. Nat Rev Mol Cell Biol 2022; 23:541-558. [PMID: 35383336 DOI: 10.1038/s41580-022-00473-y] [Citation(s) in RCA: 97] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/07/2022] [Indexed: 02/08/2023]
Abstract
Microtubules are polarized cytoskeletal filaments that serve as tracks for intracellular transport and form a scaffold that positions organelles and other cellular components and modulates cell shape and mechanics. In animal cells, the geometry, density and directionality of microtubule networks are major determinants of cellular architecture, polarity and proliferation. In dividing cells, microtubules form bipolar spindles that pull chromosomes apart, whereas in interphase cells, microtubules are organized in a cell type-specific fashion, which strongly correlates with cell physiology. In motile cells, such as fibroblasts and immune cells, microtubules are organized as radial asters, whereas in immotile epithelial and neuronal cells and in muscles, microtubules form parallel or antiparallel arrays and cortical meshworks. Here, we review recent work addressing how the formation of such microtubule networks is driven by the plethora of microtubule regulatory proteins. These include proteins that nucleate or anchor microtubule ends at different cellular structures and those that sever or move microtubules, as well as regulators of microtubule elongation, stability, bundling or modifications. The emerging picture, although still very incomplete, shows a remarkable diversity of cell-specific mechanisms that employ conserved building blocks to adjust microtubule organization in order to facilitate different cellular functions.
Collapse
Affiliation(s)
- Anna Akhmanova
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Utrecht, the Netherlands.
| | - Lukas C Kapitein
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Utrecht, the Netherlands.
| |
Collapse
|
13
|
Komatsu M, Nakamura K, Takeda T, Chiwaki F, Banno K, Aoki D, Takeshita F, Sasaki H. Aurora kinase blockade drives de novo addiction of cervical squamous cell carcinoma to druggable EGFR signalling. Oncogene 2022; 41:2326-2339. [DOI: 10.1038/s41388-022-02256-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 02/08/2022] [Accepted: 02/17/2022] [Indexed: 12/24/2022]
|
14
|
Klebanovych A, Vinopal S, Dráberová E, Sládková V, Sulimenko T, Sulimenko V, Vosecká V, Macůrek L, Legido A, Dráber P. C53 Interacting with UFM1-Protein Ligase 1 Regulates Microtubule Nucleation in Response to ER Stress. Cells 2022; 11:cells11030555. [PMID: 35159364 PMCID: PMC8834445 DOI: 10.3390/cells11030555] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 01/27/2022] [Accepted: 02/03/2022] [Indexed: 02/01/2023] Open
Abstract
ER distribution depends on microtubules, and ER homeostasis disturbance activates the unfolded protein response resulting in ER remodeling. CDK5RAP3 (C53) implicated in various signaling pathways interacts with UFM1-protein ligase 1 (UFL1), which mediates the ufmylation of proteins in response to ER stress. Here we find that UFL1 and C53 associate with γ-tubulin ring complex proteins. Knockout of UFL1 or C53 in human osteosarcoma cells induces ER stress and boosts centrosomal microtubule nucleation accompanied by γ-tubulin accumulation, microtubule formation, and ER expansion. C53, which is stabilized by UFL1, associates with the centrosome and rescues microtubule nucleation in cells lacking UFL1. Pharmacological induction of ER stress by tunicamycin also leads to increased microtubule nucleation and ER expansion. Furthermore, tunicamycin suppresses the association of C53 with the centrosome. These findings point to a novel mechanism for the relief of ER stress by stimulation of centrosomal microtubule nucleation.
Collapse
Affiliation(s)
- Anastasiya Klebanovych
- Laboratory of Biology of Cytoskeleton, Institute of Molecular Genetics of the Czech Academy of Sciences, CZ 142 20 Prague, Czech Republic; (A.K.); (S.V.); (E.D.); (V.S.); (T.S.); (V.S.); (V.V.); (L.M.)
| | - Stanislav Vinopal
- Laboratory of Biology of Cytoskeleton, Institute of Molecular Genetics of the Czech Academy of Sciences, CZ 142 20 Prague, Czech Republic; (A.K.); (S.V.); (E.D.); (V.S.); (T.S.); (V.S.); (V.V.); (L.M.)
| | - Eduarda Dráberová
- Laboratory of Biology of Cytoskeleton, Institute of Molecular Genetics of the Czech Academy of Sciences, CZ 142 20 Prague, Czech Republic; (A.K.); (S.V.); (E.D.); (V.S.); (T.S.); (V.S.); (V.V.); (L.M.)
| | - Vladimíra Sládková
- Laboratory of Biology of Cytoskeleton, Institute of Molecular Genetics of the Czech Academy of Sciences, CZ 142 20 Prague, Czech Republic; (A.K.); (S.V.); (E.D.); (V.S.); (T.S.); (V.S.); (V.V.); (L.M.)
| | - Tetyana Sulimenko
- Laboratory of Biology of Cytoskeleton, Institute of Molecular Genetics of the Czech Academy of Sciences, CZ 142 20 Prague, Czech Republic; (A.K.); (S.V.); (E.D.); (V.S.); (T.S.); (V.S.); (V.V.); (L.M.)
| | - Vadym Sulimenko
- Laboratory of Biology of Cytoskeleton, Institute of Molecular Genetics of the Czech Academy of Sciences, CZ 142 20 Prague, Czech Republic; (A.K.); (S.V.); (E.D.); (V.S.); (T.S.); (V.S.); (V.V.); (L.M.)
| | - Věra Vosecká
- Laboratory of Biology of Cytoskeleton, Institute of Molecular Genetics of the Czech Academy of Sciences, CZ 142 20 Prague, Czech Republic; (A.K.); (S.V.); (E.D.); (V.S.); (T.S.); (V.S.); (V.V.); (L.M.)
| | - Libor Macůrek
- Laboratory of Biology of Cytoskeleton, Institute of Molecular Genetics of the Czech Academy of Sciences, CZ 142 20 Prague, Czech Republic; (A.K.); (S.V.); (E.D.); (V.S.); (T.S.); (V.S.); (V.V.); (L.M.)
| | - Agustin Legido
- Section of Neurology, St. Christopher’s Hospital for Children, Department of Pediatrics, Drexel University College of Medicine, Philadelphia, PA 19134, USA;
| | - Pavel Dráber
- Laboratory of Biology of Cytoskeleton, Institute of Molecular Genetics of the Czech Academy of Sciences, CZ 142 20 Prague, Czech Republic; (A.K.); (S.V.); (E.D.); (V.S.); (T.S.); (V.S.); (V.V.); (L.M.)
- Correspondence: ; Tel.: +420-241-062-632
| |
Collapse
|
15
|
Noureddine A, Paffett ML, Franco S, Chan AE, Pallikkuth S, Lidke K, Serda RE. Endolysosomal Mesoporous Silica Nanoparticle Trafficking along Microtubular Highways. Pharmaceutics 2021; 14:pharmaceutics14010056. [PMID: 35056951 PMCID: PMC8781846 DOI: 10.3390/pharmaceutics14010056] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 12/19/2021] [Accepted: 12/21/2021] [Indexed: 11/16/2022] Open
Abstract
This study examines intra- and intercellular trafficking of mesoporous silica nanoparticles along microtubular highways, with an emphasis on intercellular bridges connecting interphase and telophase cells. The study of nanoparticle trafficking within and between cells during all phases of the cell cycle is relevant to payload destination and dilution, and impacts delivery of therapeutic or diagnostic agents. Super-resolution stochastic optical reconstruction and sub-airy unit image acquisition, the latter combined with Huygens deconvolution microscopy, enable single nanoparticle and microtubule resolution. Combined structural and functional data provide enhanced details on biological processes, with an example of mitotic inheritance during cancer cell trivision.
Collapse
Affiliation(s)
- Achraf Noureddine
- Department of Chemical and Biological Engineering, University of New Mexico, Albuquerque, NM 87131, USA;
| | - Michael L. Paffett
- Fluorescence Microscopy Shared Resource, University of New Mexico Comprehensive Cancer Center, Albuquerque, NM 87131, USA;
| | - Stefan Franco
- Internal Medicine, University of New Mexico Health Science Center, Albuquerque, NM 87131, USA; (S.F.); (A.E.C.)
| | - Alfonso E. Chan
- Internal Medicine, University of New Mexico Health Science Center, Albuquerque, NM 87131, USA; (S.F.); (A.E.C.)
| | - Sandeep Pallikkuth
- Physics and Astronomy, University of New Mexico, Albuquerque, NM 87131, USA; (S.P.); (K.L.)
| | - Keith Lidke
- Physics and Astronomy, University of New Mexico, Albuquerque, NM 87131, USA; (S.P.); (K.L.)
| | - Rita E. Serda
- Department of Chemical and Biological Engineering, University of New Mexico, Albuquerque, NM 87131, USA;
- Internal Medicine, University of New Mexico Health Science Center, Albuquerque, NM 87131, USA; (S.F.); (A.E.C.)
- Correspondence: ; Tel.: +1-505-272-7698
| |
Collapse
|
16
|
Dráber P, Dráberová E. Dysregulation of Microtubule Nucleating Proteins in Cancer Cells. Cancers (Basel) 2021; 13:cancers13225638. [PMID: 34830792 PMCID: PMC8616210 DOI: 10.3390/cancers13225638] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 11/06/2021] [Accepted: 11/08/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary The dysfunction of microtubule nucleation in cancer cells changes the overall cytoskeleton organization and cellular physiology. This review focuses on the dysregulation of the γ-tubulin ring complex (γ-TuRC) proteins that are essential for microtubule nucleation. Recent research on the high-resolution structure of γ-TuRC has brought new insight into the microtubule nucleation mechanism. We discuss the effect of γ-TuRC protein overexpression on cancer cell behavior and new drugs directed to γ-tubulin that may offer a viable alternative to microtubule-targeting agents currently used in cancer chemotherapy. Abstract In cells, microtubules typically nucleate from microtubule organizing centers, such as centrosomes. γ-Tubulin, which forms multiprotein complexes, is essential for nucleation. The γ-tubulin ring complex (γ-TuRC) is an efficient microtubule nucleator that requires additional centrosomal proteins for its activation and targeting. Evidence suggests that there is a dysfunction of centrosomal microtubule nucleation in cancer cells. Despite decades of molecular analysis of γ-TuRC and its interacting factors, the mechanisms of microtubule nucleation in normal and cancer cells remains obscure. Here, we review recent work on the high-resolution structure of γ-TuRC, which brings new insight into the mechanism of microtubule nucleation. We discuss the effects of γ-TuRC protein dysregulation on cancer cell behavior and new compounds targeting γ-tubulin. Drugs inhibiting γ-TuRC functions could represent an alternative to microtubule targeting agents in cancer chemotherapy.
Collapse
|
17
|
Sanchez AD, Branon TC, Cote LE, Papagiannakis A, Liang X, Pickett MA, Shen K, Jacobs-Wagner C, Ting AY, Feldman JL. Proximity labeling reveals non-centrosomal microtubule-organizing center components required for microtubule growth and localization. Curr Biol 2021; 31:3586-3600.e11. [PMID: 34242576 PMCID: PMC8478408 DOI: 10.1016/j.cub.2021.06.021] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 04/13/2021] [Accepted: 06/09/2021] [Indexed: 12/24/2022]
Abstract
Microtubules are polarized intracellular polymers that play key roles in the cell, including in transport, polarity, and cell division. Across eukaryotic cell types, microtubules adopt diverse intracellular organization to accommodate these distinct functions coordinated by specific cellular sites called microtubule-organizing centers (MTOCs). Over 50 years of research on MTOC biology has focused mainly on the centrosome; however, most differentiated cells employ non-centrosomal MTOCs (ncMTOCs) to organize their microtubules into diverse arrays, which are critical to cell function. To identify essential ncMTOC components, we developed the biotin ligase-based, proximity-labeling approach TurboID for use in C. elegans. We identified proteins proximal to the microtubule minus end protein PTRN-1/Patronin at the apical ncMTOC of intestinal epithelial cells, focusing on two conserved proteins: spectraplakin protein VAB-10B/MACF1 and WDR-62, a protein we identify as homologous to vertebrate primary microcephaly disease protein WDR62. VAB-10B and WDR-62 do not associate with the centrosome and instead specifically regulate non-centrosomal microtubules and the apical targeting of microtubule minus-end proteins. Depletion of VAB-10B resulted in microtubule mislocalization and delayed localization of a microtubule nucleation complex ɣ-tubulin ring complex (γ-TuRC), while loss of WDR-62 decreased the number of dynamic microtubules and abolished γ-TuRC localization. This regulation occurs downstream of cell polarity and in conjunction with actin. As this is the first report for non-centrosomal roles of WDR62 family proteins, we expand the basic cell biological roles of this important disease protein. Our studies identify essential ncMTOC components and suggest a division of labor where microtubule growth and localization are distinctly regulated.
Collapse
Affiliation(s)
- Ariana D Sanchez
- Department of Biology, Stanford University, 371 Serra Mall, Stanford, CA 94305, USA
| | - Tess C Branon
- Department of Biology, Stanford University, 371 Serra Mall, Stanford, CA 94305, USA; Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA; Departments of Genetics and Chemistry, Stanford University, Stanford, CA, USA
| | - Lauren E Cote
- Department of Biology, Stanford University, 371 Serra Mall, Stanford, CA 94305, USA
| | | | - Xing Liang
- Department of Biology, Stanford University, 371 Serra Mall, Stanford, CA 94305, USA; Howard Hughes Medical Institute, Stanford University, Stanford, CA, USA
| | - Melissa A Pickett
- Department of Biology, Stanford University, 371 Serra Mall, Stanford, CA 94305, USA
| | - Kang Shen
- Department of Biology, Stanford University, 371 Serra Mall, Stanford, CA 94305, USA; Howard Hughes Medical Institute, Stanford University, Stanford, CA, USA
| | - Christine Jacobs-Wagner
- Howard Hughes Medical Institute, Stanford University, Stanford, CA, USA; Department of Biology and ChEM-H Institute, Stanford University, Stanford, CA, USA
| | - Alice Y Ting
- Department of Biology, Stanford University, 371 Serra Mall, Stanford, CA 94305, USA; Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA; Departments of Genetics and Chemistry, Stanford University, Stanford, CA, USA; Chan Zuckerberg Biohub, San Francisco, CA, USA
| | - Jessica L Feldman
- Department of Biology, Stanford University, 371 Serra Mall, Stanford, CA 94305, USA.
| |
Collapse
|
18
|
CD13 orients the apical-basal polarity axis necessary for lumen formation. Nat Commun 2021; 12:4697. [PMID: 34349123 PMCID: PMC8338993 DOI: 10.1038/s41467-021-24993-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Accepted: 07/09/2021] [Indexed: 02/07/2023] Open
Abstract
Polarized epithelial cells can organize into complex structures with a characteristic central lumen. Lumen formation requires that cells coordinately orient their polarity axis so that the basolateral domain is on the outside and apical domain inside epithelial structures. Here we show that the transmembrane aminopeptidase, CD13, is a key determinant of epithelial polarity orientation. CD13 localizes to the apical membrane and associates with an apical complex with Par6. CD13-deficient cells display inverted polarity in which apical proteins are retained on the outer cell periphery and fail to accumulate at an intercellular apical initiation site. Here we show that CD13 is required to couple apical protein cargo to Rab11-endosomes and for capture of endosomes at the apical initiation site. This role in polarity utilizes the short intracellular domain but is independent of CD13 peptidase activity.
Collapse
|
19
|
The γ-tubulin meshwork assists in the recruitment of PCNA to chromatin in mammalian cells. Commun Biol 2021; 4:767. [PMID: 34158617 PMCID: PMC8219688 DOI: 10.1038/s42003-021-02280-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Accepted: 05/28/2021] [Indexed: 12/27/2022] Open
Abstract
Changes in the location of γ-tubulin ensure cell survival and preserve genome integrity. We investigated whether the nuclear accumulation of γ-tubulin facilitates the transport of proliferating cell nuclear antigen (PCNA) between the cytosolic and the nuclear compartment in mammalian cells. We found that the γ-tubulin meshwork assists in the recruitment of PCNA to chromatin. Also, decreased levels of γ-tubulin reduce the nuclear pool of PCNA. In addition, the γ-tubulin C terminus encodes a PCNA-interacting peptide (PIP) motif, and a γ-tubulin–PIP-mutant affects the nuclear accumulation of PCNA. In a cell-free system, PCNA and γ-tubulin formed a complex. In tumors, there is a significant positive correlation between TUBG1 and PCNA expression. Thus, we report a novel mechanism that constitutes the basis for tumor growth by which the γ-tubulin meshwork maintains indefinite proliferation by acting as an opportune scaffold for the transport of PCNA from the cytosol to the chromatin. Corvaisier et al discover that γ-tubulin and replication protein PCNA forms a complex and that this facilitates recruitment of PCNA to chromatin both during cell division and during the DSB repair response. They identify a PCNA binding motif in γ-tubulin, which when mutated affects replication fork progression, providing insights into the role of the nuclear γ-tubulin meshwork.
Collapse
|
20
|
Lattao R, Rangone H, Llamazares S, Glover DM. Mauve/LYST limits fusion of lysosome-related organelles and promotes centrosomal recruitment of microtubule nucleating proteins. Dev Cell 2021; 56:1000-1013.e6. [PMID: 33725482 PMCID: PMC8024676 DOI: 10.1016/j.devcel.2021.02.019] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 11/17/2020] [Accepted: 02/17/2021] [Indexed: 11/28/2022]
Abstract
Lysosome-related organelles (LROs) are endosomal compartments carrying tissue-specific proteins, which become enlarged in Chediak-Higashi syndrome (CHS) due to mutations in LYST. Here, we show that Drosophila Mauve, a counterpart of LYST, suppresses vesicle fusion events with lipid droplets (LDs) during the formation of yolk granules (YGs), the LROs of the syncytial embryo, and opposes Rab5, which promotes fusion. Mauve localizes on YGs and at spindle poles, and it co-immunoprecipitates with the LDs' component and microtubule-associated protein Minispindles/Ch-TOG. Minispindles levels are increased at the enlarged YGs and diminished around centrosomes in mauve-derived mutant embryos. This leads to decreased microtubule nucleation from centrosomes, a defect that can be rescued by dominant-negative Rab5. Together, this reveals an unanticipated link between endosomal vesicles and centrosomes. These findings establish Mauve/LYST's role in regulating LRO formation and centrosome behavior, a role that could account for the enlarged LROs and centrosome positioning defects at the immune synapse of CHS patients.
Collapse
Affiliation(s)
- Ramona Lattao
- University of Cambridge, Department of Genetics, Downing Street, Cambridge CB23EH, UK.
| | - Hélène Rangone
- University of Cambridge, Department of Genetics, Downing Street, Cambridge CB23EH, UK
| | - Salud Llamazares
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Parc Cientific de Barcelona, C/ Baldiri Reixac 10, 08028 Barcelona, Spain
| | - David M Glover
- University of Cambridge, Department of Genetics, Downing Street, Cambridge CB23EH, UK; Division of Biology and Biological Engineering, California Institute of Technology, 1200 E, California Blvd, Pasadena, CA 91125, USA.
| |
Collapse
|
21
|
Willoughby PM, Allen M, Yu J, Korytnikov R, Chen T, Liu Y, So I, Macpherson N, Mitchell JA, Fernandez-Gonzalez R, Bruce AE. The recycling endosome protein Rab25 coordinates collective cell movements in the zebrafish surface epithelium. eLife 2021; 10:66060. [PMID: 33755014 PMCID: PMC8034978 DOI: 10.7554/elife.66060] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 03/22/2021] [Indexed: 12/16/2022] Open
Abstract
In emerging epithelial tissues, cells undergo dramatic rearrangements to promote tissue shape changes. Dividing cells remain interconnected via transient cytokinetic bridges. Bridges are cleaved during abscission and currently, the consequences of disrupting abscission in developing epithelia are not well understood. We show that the Rab GTPase Rab25 localizes near cytokinetic midbodies and likely coordinates abscission through endomembrane trafficking in the epithelium of the zebrafish gastrula during epiboly. In maternal-zygotic Rab25a and Rab25b mutant embryos, morphogenic activity tears open persistent apical cytokinetic bridges that failed to undergo timely abscission. Cytokinesis defects result in anisotropic cell morphologies that are associated with a reduction of contractile actomyosin networks. This slows cell rearrangements and alters the viscoelastic responses of the tissue, all of which likely contribute to delayed epiboly. We present a model in which Rab25 trafficking coordinates cytokinetic bridge abscission and cortical actin density, impacting local cell shape changes and tissue-scale forces.
Collapse
Affiliation(s)
| | - Molly Allen
- Department of Cell and Systems Biology, University of Toronto, Toronto, Canada
| | - Jessica Yu
- Ted Rogers Centre for Heart Research, Translational Biology and Engineering Program, University of Toronto, Toronto, Canada.,Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Canada
| | - Roman Korytnikov
- Department of Cell and Systems Biology, University of Toronto, Toronto, Canada
| | - Tianhui Chen
- Department of Cell and Systems Biology, University of Toronto, Toronto, Canada
| | - Yupeng Liu
- Department of Cell and Systems Biology, University of Toronto, Toronto, Canada
| | - Isis So
- Department of Cell and Systems Biology, University of Toronto, Toronto, Canada
| | - Neil Macpherson
- Department of Cell and Systems Biology, University of Toronto, Toronto, Canada
| | - Jennifer A Mitchell
- Department of Cell and Systems Biology, University of Toronto, Toronto, Canada
| | - Rodrigo Fernandez-Gonzalez
- Department of Cell and Systems Biology, University of Toronto, Toronto, Canada.,Ted Rogers Centre for Heart Research, Translational Biology and Engineering Program, University of Toronto, Toronto, Canada.,Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Canada
| | - Ashley Ee Bruce
- Department of Cell and Systems Biology, University of Toronto, Toronto, Canada
| |
Collapse
|
22
|
To nucleate or not, that is the question in neurons. Neurosci Lett 2021; 751:135806. [PMID: 33705928 DOI: 10.1016/j.neulet.2021.135806] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 02/23/2021] [Accepted: 02/26/2021] [Indexed: 01/19/2023]
Abstract
Microtubules are the structural center of neurons, stretching in overlapping arrays from the cell body to the far reaches of axons and dendrites. They also act as the tracks for long-range transport mediated by dynein and kinesin motors. Transcription and most translation take place in the cell body, and newly made cargoes must be shipped from this site of synthesis to sites of function in axons and dendrites. This constant demand for transport means that the microtubule array must be present without gaps throughout the cell over the lifetime of the animal. This task is made slightly easier in many animals by the relatively long, stable microtubules present in neurons. However, even stable neuronal microtubules have ends that are dynamic, and individual microtubules typically last on the order of hours, while the neurons around them last a lifetime. "Birth" of new microtubules is therefore required to maintain the neuronal microtubule array. In this review we discuss the nucleation of new microtubules in axons and dendrites, including how and where they are nucleated. In addition, it is becoming clear that neuronal microtubule nucleation is highly regulated, with unexpected machinery impinging on the decision of whether nucleation sites are active or inactive through space and time.
Collapse
|
23
|
Guo H, Wei JH, Zhang Y, Seemann J. Importin α phosphorylation promotes TPX2 activation by GM130 to control astral microtubules and spindle orientation. J Cell Sci 2021; 134:jcs.258356. [PMID: 33526712 DOI: 10.1242/jcs.258356] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2020] [Accepted: 01/11/2021] [Indexed: 01/10/2023] Open
Abstract
Spindle orientation is important in multiple developmental processes as it determines cell fate and function. The orientation of the spindle depends on the assembly of a proper astral microtubule network. Here, we report that the spindle assembly factor TPX2 regulates astral microtubules. TPX2 in the spindle pole area is activated by GM130 (GOLGA2) on Golgi membranes to promote astral microtubule growth. GM130 relieves TPX2 inhibition by competing for importin α1 (KPNA2) binding. Mitotic phosphorylation of importin α at serine 62 (S62) by CDK1 switches its substrate preference from TPX2 to GM130, thereby enabling competition-based activation. Importin α S62A mutation impedes local TPX2 activation and compromises astral microtubule formation, ultimately resulting in misoriented spindles. Blocking the GM130-importin α-TPX2 pathway impairs astral microtubule growth. Our results reveal a novel role for TPX2 in the organization of astral microtubules. Furthermore, we show that the substrate preference of the important mitotic modulator importin α is regulated by CDK1-mediated phosphorylation.
Collapse
Affiliation(s)
- Haijing Guo
- Department of Cell Biology, University of Texas Southwestern Medical Center, 6000 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Jen-Hsuan Wei
- Institute of Molecular Biology, Academia Sinica, Taipei 11529, Taiwan
| | - Yijun Zhang
- Department of Cell Biology, University of Texas Southwestern Medical Center, 6000 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Joachim Seemann
- Department of Cell Biology, University of Texas Southwestern Medical Center, 6000 Harry Hines Boulevard, Dallas, TX 75390, USA
| |
Collapse
|
24
|
Hall NA, Hehnly H. A centriole's subdistal appendages: contributions to cell division, ciliogenesis and differentiation. Open Biol 2021; 11:200399. [PMID: 33561384 PMCID: PMC8061701 DOI: 10.1098/rsob.200399] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The centrosome is a highly conserved structure composed of two centrioles surrounded by pericentriolar material. The mother, and inherently older, centriole has distal and subdistal appendages, whereas the daughter centriole is devoid of these appendage structures. Both appendages have been primarily linked to functions in cilia formation. However, subdistal appendages present with a variety of potential functions that include spindle placement, chromosome alignment, the final stage of cell division (abscission) and potentially cell differentiation. Subdistal appendages are particularly interesting in that they do not always display a conserved ninefold symmetry in appendage organization on the mother centriole across eukaryotic species, unlike distal appendages. In this review, we aim to differentiate both the morphology and role of the distal and subdistal appendages, with a particular focus on subdistal appendages.
Collapse
Affiliation(s)
- Nicole A Hall
- Department of Biology, Syracuse University, Syracuse NY, USA
| | - Heidi Hehnly
- Department of Biology, Syracuse University, Syracuse NY, USA
| |
Collapse
|
25
|
Promotion of cancer cell stemness by Ras. Biochem Soc Trans 2021; 49:467-476. [PMID: 33544116 PMCID: PMC7925005 DOI: 10.1042/bst20200964] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 01/17/2021] [Accepted: 01/19/2021] [Indexed: 02/07/2023]
Abstract
Cancer stem cells (CSC) may be the most relevant and elusive cancer cell population, as they have the exquisite ability to seed new tumors. It is plausible, that highly mutated cancer genes, such as KRAS, are functionally associated with processes contributing to the emergence of stemness traits. In this review, we will summarize the evidence for a stemness driving activity of oncogenic Ras. This activity appears to differ by Ras isoform, with the highly mutated KRAS having a particularly profound impact. Next to established stemness pathways such as Wnt and Hedgehog (Hh), the precise, cell cycle dependent orchestration of the MAPK-pathway appears to relay Ras activation in this context. We will examine how non-canonical activities of K-Ras4B (hereafter K-Ras) could be enabled by its trafficking chaperones calmodulin and PDE6D/PDEδ. Both dynamically localize to the cellular machinery that is intimately linked to cell fate decisions, such as the primary cilium and the centrosome. Thus, it can be speculated that oncogenic K-Ras disrupts fundamental polarized signaling and asymmetric apportioning processes that are necessary during cell differentiation.
Collapse
|
26
|
Cyclin-dependent Kinase 1 and Aurora Kinase choreograph mitotic storage and redistribution of a growth factor receptor. PLoS Biol 2021; 19:e3001029. [PMID: 33395410 PMCID: PMC7808676 DOI: 10.1371/journal.pbio.3001029] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 01/14/2021] [Accepted: 12/18/2020] [Indexed: 01/08/2023] Open
Abstract
Endosomal trafficking of receptors and associated proteins plays a critical role in signal processing. Until recently, it was thought that trafficking was shut down during cell division. Thus, remarkably, the regulation of trafficking during division remains poorly characterized. Here we delineate the role of mitotic kinases in receptor trafficking during asymmetric division. Targeted perturbations reveal that Cyclin-dependent Kinase 1 (CDK1) and Aurora Kinase promote storage of Fibroblast Growth Factor Receptors (FGFRs) by suppressing endosomal degradation and recycling pathways. As cells progress through metaphase, loss of CDK1 activity permits differential degradation and targeted recycling of stored receptors, leading to asymmetric induction. Mitotic receptor storage, as delineated in this study, may facilitate rapid reestablishment of signaling competence in nascent daughter cells. However, mutations that limit or enhance the release of stored signaling components could alter daughter cell fate or behavior thereby promoting oncogenesis. This study provides fundamental insights into the crosstalk between cell division and signaling, with implications for cancer. High-resolution in vivo analysis reveals that dividing cells sequester signal receptor proteins into internal compartments; stored receptors are then redistributed as cells complete division.
Collapse
|
27
|
Rab11a Is Overexpressed in Gastric Cancer and Regulates FAK/AKT Signaling. JOURNAL OF ONCOLOGY 2020; 2020:3494396. [PMID: 33178272 PMCID: PMC7648696 DOI: 10.1155/2020/3494396] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 09/15/2020] [Accepted: 09/28/2020] [Indexed: 02/06/2023]
Abstract
Dysregulation of Rab11a has been implicated in the progression of several cancers. However, there have been no such studies for human gastric cancers. In the current study, we examined Rab11a protein expression and found it was upregulated in 49 of 108 gastric cancer tissues and correlated with local invasion, nodal metastasis, and advanced stage. Rab11a protein was higher in gastric cancer cell lines than normal gastric cell line. We transfected Rab11a plasmid and siRNA in both MGC803 and AGS cell lines. Rab11a overexpression increased the cell growth rate, colony numbers, and invasion ability in both MGC803 and AGS cell lines. Downregulation of Rab11a using siRNA decreased the cell proliferation rate, colony numbers, and inhibited invasion. Rab11a overexpression also conferred cisplatin resistance. Annexin V/PI staining showed that Rab11a overexpression suppressed cisplatin-induced apoptosis, while Rab11a depletion promoted cell apoptosis. We also showed that Rab11a overexpression maintained mitochondrial membrane potential. Western blot analysis revealed that Rab11a increased protein expression of MMP2, cyclin D1, Bcl-2, p-FAK, and p-AKT, while Rab11a depletion showed the opposite effects. Blockage of FAK using inhibitor downregulated Bcl-2, cyclin D1, MMP2, and p-AKT expression and abolished the effects of Rab11a on these proteins. In summary, our data demonstrated that Rab11a is upregulated in human gastric cancers. Rab11a facilitated cell proliferation and invasion, as well as cisplatin sensitivity and mitochondrial membrane potential, possibly via the FAK/AKT signaling pathway.
Collapse
|
28
|
Corvaisier M, Alvarado-Kristensson M. Non-Canonical Functions of the Gamma-Tubulin Meshwork in the Regulation of the Nuclear Architecture. Cancers (Basel) 2020; 12:cancers12113102. [PMID: 33114224 PMCID: PMC7690915 DOI: 10.3390/cancers12113102] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 10/17/2020] [Accepted: 10/21/2020] [Indexed: 12/15/2022] Open
Abstract
Simple Summary The appearance of a cell is connected to its function. For example, the fusiform of smooth muscle cells is adapted to facilitate muscle contraction, the lobed nucleus in white blood cells assists with the migratory behavior of these immune cells, and the condensed nucleus in sperm aids in their swimming efficiency. Thus, changes in appearance have been used for decades by doctors as a diagnostic method for human cancers. Here, we summarize our knowledge of how a cell maintains the shape of the nuclear compartment. Specifically, we discuss the role of a novel protein meshwork, the gamma-tubulin meshwork, in the regulation of nuclear morphology and as a therapeutic target against cancer. Abstract The nuclear architecture describes the organization of the various compartments in the nucleus of eukaryotic cells, where a plethora of processes such as nucleocytoplasmic transport, gene expression, and assembly of ribosomal subunits occur in a dynamic manner. During the different phases of the cell cycle, in post-mitotic cells and after oncogenic transformation, rearrangements of the nuclear architecture take place, and, among other things, these alterations result in reorganization of the chromatin and changes in gene expression. A member of the tubulin family, γtubulin, was first identified as part of a multiprotein complex that allows nucleation of microtubules. However, more than a decade ago, γtubulin was also characterized as a nuclear protein that modulates several crucial processes that affect the architecture of the nucleus. This review presents the latest knowledge regarding changes that arise in the nuclear architecture of healthy cells and under pathological conditions and, more specifically, considers the particular involvement of γtubulin in the modulation of the biology of the nuclear compartment.
Collapse
|
29
|
Liang X, Kokes M, Fetter RD, Sallee MD, Moore AW, Feldman JL, Shen K. Growth cone-localized microtubule organizing center establishes microtubule orientation in dendrites. eLife 2020; 9:e56547. [PMID: 32657271 PMCID: PMC7375809 DOI: 10.7554/elife.56547] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 07/09/2020] [Indexed: 01/01/2023] Open
Abstract
A polarized arrangement of neuronal microtubule arrays is the foundation of membrane trafficking and subcellular compartmentalization. Conserved among both invertebrates and vertebrates, axons contain exclusively 'plus-end-out' microtubules while dendrites contain a high percentage of 'minus-end-out' microtubules, the origins of which have been a mystery. Here we show that in Caenorhabditis elegans the dendritic growth cone contains a non-centrosomal microtubule organizing center (MTOC), which generates minus-end-out microtubules along outgrowing dendrites and plus-end-out microtubules in the growth cone. RAB-11-positive endosomes accumulate in this region and co-migrate with the microtubule nucleation complex γ-TuRC. The MTOC tracks the extending growth cone by kinesin-1/UNC-116-mediated endosome movements on distal plus-end-out microtubules and dynein clusters this advancing MTOC. Critically, perturbation of the function or localization of the MTOC causes reversed microtubule polarity in dendrites. These findings unveil the endosome-localized dendritic MTOC as a critical organelle for establishing axon-dendrite polarity.
Collapse
Affiliation(s)
- Xing Liang
- Department of Biology, Stanford UniversityStanfordUnited States
- Howard Hughes Medical Institute, Stanford UniversityStanfordUnited States
| | - Marcela Kokes
- Department of Biology, Stanford UniversityStanfordUnited States
- Howard Hughes Medical Institute, Stanford UniversityStanfordUnited States
| | - Richard D Fetter
- Howard Hughes Medical Institute, Stanford UniversityStanfordUnited States
| | | | | | | | - Kang Shen
- Department of Biology, Stanford UniversityStanfordUnited States
- Howard Hughes Medical Institute, Stanford UniversityStanfordUnited States
| |
Collapse
|
30
|
Hwang JH, Vuong LT, Choi KW. Crumbs, Galla and Xpd are required for Kinesin-5 regulation in mitosis and organ growth in Drosophila. J Cell Sci 2020; 133:jcs246801. [PMID: 32501288 DOI: 10.1242/jcs.246801] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Accepted: 04/29/2020] [Indexed: 12/20/2022] Open
Abstract
Xeroderma Pigmentosum D (XPD, also known as ERCC2) is a multi-functional protein involved in transcription, DNA repair and chromosome segregation. In Drosophila, Xpd interacts with Crumbs (Crb) and Galla to regulate mitosis during embryogenesis. It is unknown how these proteins are linked to mitosis. Here, we show that Crb, Galla-2 and Xpd regulate nuclear division in the syncytial embryo by interacting with Klp61F, the Drosophila mitotic Kinesin-5 associated with bipolar spindles. Crb, Galla-2 and Xpd physically interact with Klp61F and colocalize to mitotic spindles. Knockdown of any of these proteins results in similar mitotic defects. These phenotypes are restored by overexpression of Klp61F, suggesting that Klp61F is a major effector. Mitotic defects of galla-2 RNAi are suppressed by Xpd overexpression but not vice versa. Depletion of Crb, Galla-2 or Xpd results in a reduction of Klp61F levels. Reducing proteasome function restores Klp61F levels and suppresses mitotic defects caused by knockdown of Crb, Galla-2 or Xpd. Furthermore, eye growth is regulated by Xpd and Klp61F. Hence, we propose that Crb, Galla-2 and Xpd interact to maintain the level of Klp61F during mitosis and organ growth.
Collapse
Affiliation(s)
- Ji-Hyun Hwang
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 34141, Korea
| | - Linh Thuong Vuong
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 34141, Korea
| | - Kwang-Wook Choi
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 34141, Korea
| |
Collapse
|
31
|
Sharma A, Dagar S, Mylavarapu SVS. Transgelin-2 and phosphoregulation of the LIC2 subunit of dynein govern mitotic spindle orientation. J Cell Sci 2020; 133:jcs239673. [PMID: 32467330 DOI: 10.1242/jcs.239673] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Accepted: 04/29/2020] [Indexed: 08/31/2023] Open
Abstract
The molecular motor dynein is essential for mitotic spindle orientation, which defines the axis of cell division. The light intermediate chain subunits, LIC1 and LIC2, define biochemically and functionally distinct vertebrate dynein complexes, with LIC2-dynein playing a crucial role in ensuring spindle orientation. We reveal a novel, mitosis-specific interaction of LIC2-dynein with the cortical actin-bundling protein transgelin-2. Transgelin-2 is required for maintaining proper spindle length, equatorial metaphase chromosome alignment, spindle orientation and timely anaphase onset. We show that transgelin-2 stabilizes the cortical recruitment of LGN-NuMA, which together with dynein is required for spindle orientation. The opposing actions of transgelin-2 and LIC2-dynein maintain optimal cortical levels of LGN-NuMA. In addition, we show that the highly conserved serine 194 phosphorylation of LIC2 is required for proper spindle orientation, by maintaining mitotic centrosome integrity to ensure optimal astral microtubule nucleation. The work reveals two specific mechanisms through which LIC2-dynein regulates mitotic spindle orientation; namely, through a new interactor transgelin-2, which is required for engagement of LGN-NuMA with the actin cortex, and through mitotic phosphoregulation of LIC2 to control microtubule nucleation from the poles.This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Amit Sharma
- Laboratory of Cellular Dynamics, Regional Centre for Biotechnology, NCR Biotech Science Cluster, 3rd Milestone Faridabad-Gurgaon Expressway, Faridabad, Haryana 121001, India
- Affiliated to the Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
| | - Sunayana Dagar
- Laboratory of Cellular Dynamics, Regional Centre for Biotechnology, NCR Biotech Science Cluster, 3rd Milestone Faridabad-Gurgaon Expressway, Faridabad, Haryana 121001, India
- Kalinga Institute of Industrial Technology, Bhubaneswar, Odisha 751024, India
| | - Sivaram V S Mylavarapu
- Laboratory of Cellular Dynamics, Regional Centre for Biotechnology, NCR Biotech Science Cluster, 3rd Milestone Faridabad-Gurgaon Expressway, Faridabad, Haryana 121001, India
- Affiliated to the Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
- Kalinga Institute of Industrial Technology, Bhubaneswar, Odisha 751024, India
| |
Collapse
|
32
|
Zhang ZY, Lu M, Liu ZK, Li H, Yong YL, Zhang RY, Chen ZN, Bian H. Rab11a regulates MMP2 expression by activating the PI3K/AKT pathway in human hepatocellular carcinoma cells. Pathol Res Pract 2020; 216:153046. [PMID: 32825931 DOI: 10.1016/j.prp.2020.153046] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 05/17/2020] [Accepted: 05/31/2020] [Indexed: 02/06/2023]
Abstract
As a member of the Rab GTPase family, Rab11a plays an important role in vesicle transport and tumor progression. However, it is not clear whether it can also be used as an oncoprotein in hepatocellular carcinoma (HCC). In this study, database and immunohistochemical analyses showed that Rab11a was highly expressed in HCC tissues, and associated with poor clinical prognosis. Rab11a overexpression promoted the proliferation, migration, invasion, and anti-apoptosis of human HCC cell lines, MHCC-97H and HCC-LM3, whereas the downregulation of Rab11a inhibited these biological tumor activities. Nude mice xenograft demonstrated that Rab11a had a positive effect on the growth of hepatocellular carcinoma cells in vivo. Further studies found that the PI3K/AKT pathway and matrix metalloproteinase 2 (MMP2) upregulation can be activated by over-expression of Rab11a. However, MMP2 upregulation induced by Rab11a can be inhibited by the PI3K/AKT pathway inhibitor, LY294002. Altogether, our study established for the first time that Rab11a can play a pro-cancer role in HCC, as a novel oncoprotein, by activating the PI3K/AKT pathway to regulate MMP2 expression.
Collapse
Affiliation(s)
- Zhi-Yun Zhang
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Meng Lu
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Ze-Kun Liu
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Hao Li
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Yu-Le Yong
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Ren-Yu Zhang
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Zhi-Nan Chen
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an, 710032, China.
| | - Huijie Bian
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an, 710032, China.
| |
Collapse
|
33
|
Zhang L, Xie B, Qiu Y, Jing D, Zhang J, Duan Y, Li Z, Fan M, He J, Qiu Y, Tan R, Li JJ, Sun LQ. Rab25-Mediated EGFR Recycling Causes Tumor Acquired Radioresistance. iScience 2020; 23:100997. [PMID: 32252020 PMCID: PMC7132159 DOI: 10.1016/j.isci.2020.100997] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 02/18/2020] [Accepted: 03/17/2020] [Indexed: 12/20/2022] Open
Abstract
Tumor acquired radioresistance remains as the major limit in cancer radiotherapy (RT). Rab25, a receptor recycling protein, has been reported to be enhanced in tumors with aggressive phenotype and chemotherapy resistance. In this study, elevated Rab25 expression was identified in an array of radioresistant human cancer cell lines, in vivo radioresistant xenograft tumors. Clinical investigation confirmed that Rab25 expression was also associated with a worse prognosis in patients with lung adenocarcinoma (LUAD) and nasopharyngeal carcinoma (NPC). Enhanced activities of EGFR were observed in both NPC and LUAD radioresistant cells. Rab25 interacts with EGFR to enhance EGFR recycling to cell surface and to decrease degradation in cytoplasm. Inhibition of Rab25 showed synergized radiosensitivity with reduced aggressive phenotype. This study provides the clinical and experimental evidence that Rab25 is a potential therapeutic target to alleviate the hyperactive EGFR signaling and to prevent RT-acquired tumor resistance in patients with LUAD and NPC.
Collapse
Affiliation(s)
- Lu Zhang
- Center for Molecular Medicine, Xiangya Hospital, Central South University, Changsha 410008, China; Key Laboratory of Molecular Radiation Oncology of Hunan Province, Changsha 410008, China; Department of Radiation Oncology, University of California Davis, Sacramento, CA 95817, USA
| | - Bowen Xie
- Center for Molecular Medicine, Xiangya Hospital, Central South University, Changsha 410008, China; Key Laboratory of Molecular Radiation Oncology of Hunan Province, Changsha 410008, China; Department of Radiation Oncology, University of California Davis, Sacramento, CA 95817, USA
| | - Yanfang Qiu
- Center for Molecular Medicine, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Di Jing
- Center for Molecular Medicine, Xiangya Hospital, Central South University, Changsha 410008, China; Department of Radiation Oncology, University of California Davis, Sacramento, CA 95817, USA
| | - Jing Zhang
- Center for Molecular Medicine, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Yumei Duan
- Department of Pathology, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Zhi Li
- Center for Molecular Medicine, Xiangya Hospital, Central South University, Changsha 410008, China; Key Laboratory of Molecular Radiation Oncology of Hunan Province, Changsha 410008, China
| | - Ming Fan
- Department of Radiation Oncology, University of California Davis, Sacramento, CA 95817, USA
| | - Jiang He
- Center for Molecular Medicine, Xiangya Hospital, Central South University, Changsha 410008, China; Key Laboratory of Molecular Radiation Oncology of Hunan Province, Changsha 410008, China
| | - Yuanzheng Qiu
- Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Rong Tan
- Center for Molecular Medicine, Xiangya Hospital, Central South University, Changsha 410008, China; Key Laboratory of Molecular Radiation Oncology of Hunan Province, Changsha 410008, China
| | - Jian Jian Li
- Department of Radiation Oncology, University of California Davis, Sacramento, CA 95817, USA; NCI-desginaged Comprehensive Cancer Center, Sacramento, CA 95817, USA.
| | - Lun-Quan Sun
- Center for Molecular Medicine, Xiangya Hospital, Central South University, Changsha 410008, China; Key Laboratory of Molecular Radiation Oncology of Hunan Province, Changsha 410008, China; National Clinical Research Center for Geriatric Disorders Xiangya Hospital, Changsha, China 410008.
| |
Collapse
|
34
|
Naslavsky N, Caplan S. Endocytic membrane trafficking in the control of centrosome function. Curr Opin Cell Biol 2020; 65:150-155. [PMID: 32143977 DOI: 10.1016/j.ceb.2020.01.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 01/15/2020] [Accepted: 01/17/2020] [Indexed: 12/15/2022]
Abstract
Until recently, endocytic trafficking and its regulators were thought to function almost exclusively on membrane-bound organelles and/or vesicles containing a lipid bilayer. Recent studies have demonstrated that endocytic regulatory proteins play much wider roles in trafficking regulation and influence a variety of nonendocytic pathways, including trafficking to/from mitochondria and peroxisomes. Moreover, new studies also suggest that endocytic regulators also control trafficking to and from cellular organelles that lack membranes, such as the centrosome. Although endocytic membrane trafficking (EMT) clearly impacts pathways downstream of the centrosome, such as ciliogenesis (including transport to and from cilia), mitotic spindle formation, and cytokinesis, relatively few studies have focused on the growing role for EMT more directly on centrosome biogenesis, maintenance and control throughout cell cycle, and centrosome duplication. Indeed, a growing number of endocytic regulatory proteins have been implicated in centrosome regulation, including various Rab proteins (among them Rab11) and the leucine-rich repeat kinase 2. In this review, we will examine the relationship between centrosomes and EMT, focusing primarily on how EMT directly influences the centrosome.
Collapse
Affiliation(s)
- Naava Naslavsky
- The Department of Biochemistry and Molecular Biology and Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, 68198-5870, United States
| | - Steve Caplan
- The Department of Biochemistry and Molecular Biology and Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, 68198-5870, United States.
| |
Collapse
|
35
|
Carlton JG, Jones H, Eggert US. Membrane and organelle dynamics during cell division. Nat Rev Mol Cell Biol 2020; 21:151-166. [DOI: 10.1038/s41580-019-0208-1] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/20/2019] [Indexed: 12/31/2022]
|
36
|
Targeting the IL-1β/EHD1/TUBB3 axis overcomes resistance to EGFR-TKI in NSCLC. Oncogene 2019; 39:1739-1755. [DOI: 10.1038/s41388-019-1099-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Revised: 10/29/2019] [Accepted: 11/04/2019] [Indexed: 12/24/2022]
|
37
|
Leng C, Overeem AW, Cartón-Garcia F, Li Q, Klappe K, Kuipers J, Cui Y, Zuhorn IS, Arango D, van IJzendoorn SCD. Loss of MYO5B expression deregulates late endosome size which hinders mitotic spindle orientation. PLoS Biol 2019; 17:e3000531. [PMID: 31682603 PMCID: PMC6855566 DOI: 10.1371/journal.pbio.3000531] [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: 06/25/2019] [Revised: 11/14/2019] [Accepted: 10/17/2019] [Indexed: 12/23/2022] Open
Abstract
Recycling endosomes regulate plasma membrane recycling. Recently, recycling endosome–associated proteins have been implicated in the positioning and orientation of the mitotic spindle and cytokinesis. Loss of MYO5B, encoding the recycling endosome–associated myosin Vb, is associated with tumor development and tissue architecture defects in the gastrointestinal tract. Whether loss of MYO5B expression affects mitosis is not known. Here, we demonstrate that loss of MYO5B expression delayed cytokinesis, perturbed mitotic spindle orientation, led to the misorientation of the plane of cell division during the course of mitosis, and resulted in the delamination of epithelial cells. Remarkably, the effects on spindle orientation, but not cytokinesis, were a direct consequence of physical hindrance by giant late endosomes, which were formed in a chloride channel–sensitive manner concomitant with a redistribution of chloride channels from the cell periphery to late endosomes upon loss of MYO5B. Rab7 availability was identified as a limiting factor for the development of giant late endosomes. In accordance, increasing rab7 availability corrected mitotic spindle misorientation and cell delamination in cells lacking MYO5B expression. In conclusion, we identified a novel role for MYO5B in the regulation of late endosome size control and identify the inability to control late endosome size as an unexpected novel mechanism underlying defects in cell division orientation and epithelial architecture. Loss of the recycling endosome-associated motor protein myosin Vb causes the formation of giant late endo-lysosomes; these in turn hinder the orientation of the mitotic spindle and chromosome segregation. Deregulated endosome size thus hampers faithful cell division.
Collapse
Affiliation(s)
- Changsen Leng
- Department of Biomedical Sciences of Cells and Systems, section Molecular Cell Biology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Arend W. Overeem
- Department of Biomedical Sciences of Cells and Systems, section Molecular Cell Biology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Fernando Cartón-Garcia
- Group of Biomedical Research in Digestive Tract Tumors, CIBBIM-Nanomedicine, Vall d'Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona (UAB), Barcelona, Spain
| | - Qinghong Li
- Department of Biomedical Sciences of Cells and Systems, section Molecular Cell Biology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Karin Klappe
- Department of Biomedical Sciences of Cells and Systems, section Molecular Cell Biology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Jeroen Kuipers
- Department of Biomedical Sciences of Cells and Systems, section Molecular Cell Biology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Yingying Cui
- Department of Gastroenterology and Hepatology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Inge S. Zuhorn
- Department of Biomedical Engineering, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Diego Arango
- Group of Biomedical Research in Digestive Tract Tumors, CIBBIM-Nanomedicine, Vall d'Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona (UAB), Barcelona, Spain
| | - Sven C. D. van IJzendoorn
- Department of Biomedical Sciences of Cells and Systems, section Molecular Cell Biology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
- * E-mail:
| |
Collapse
|
38
|
Centrosomal and ciliary targeting of CCDC66 requires cooperative action of centriolar satellites, microtubules and molecular motors. Sci Rep 2019; 9:14250. [PMID: 31582766 PMCID: PMC6776500 DOI: 10.1038/s41598-019-50530-4] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Accepted: 09/11/2019] [Indexed: 02/06/2023] Open
Abstract
Mammalian centrosomes and cilia play key roles in many cellular processes and their deregulation is linked to cancer and ciliopathies. Spatiotemporal regulation of their biogenesis and function in response to physiological stimuli requires timely protein targeting. This can occur by different pathways, including microtubule-dependent active transport and via centriolar satellites, which are key regulators of cilia assembly and signaling. How satellites mediate their functions and their relationship with other targeting pathways is currently unclear. To address this, we studied retinal degeneration gene product CCDC66, which localizes to centrosomes, cilia, satellites and microtubules and functions in ciliogenesis. FRAP experiments showed that its centrosomal pool was dynamic and the ciliary pool associated with the ciliary axoneme and was stable. Centrosomal CCDC66 abundance and dynamics required microtubule-dependent active transport and tethering, and was inhibited by sequestration at satellites. Systematic quantitation of satellite dynamics identified only a small fraction to display microtubule-based bimodal motility, consistent with trafficking function. Majority displayed diffusive motility with unimodal persistence, supporting sequestration function. Together, our findings reveal new mechanisms of communication between membrane-less compartments.
Collapse
|
39
|
Karabasheva D, Smyth JT. A novel, dynein-independent mechanism focuses the endoplasmic reticulum around spindle poles in dividing Drosophila spermatocytes. Sci Rep 2019; 9:12456. [PMID: 31462700 PMCID: PMC6713755 DOI: 10.1038/s41598-019-48860-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Accepted: 08/14/2019] [Indexed: 01/04/2023] Open
Abstract
In dividing animal cells the endoplasmic reticulum (ER) concentrates around the poles of the spindle apparatus by associating with astral microtubules (MTs), and this association is essential for proper ER partitioning to progeny cells. The mechanisms that associate the ER with astral MTs are unknown. Because astral MT minus-ends are anchored by centrosomes at spindle poles, we hypothesized that the MT minus-end motor dynein mediates ER concentration around spindle poles. Live in vivo imaging of Drosophila spermatocytes revealed that dynein is required for ER concentration around centrosomes during late interphase. In marked contrast, however, dynein suppression had no effect on ER association with astral MTs and concentration around spindle poles in early M-phase. In fact, there was a sudden onset of ER association with astral MTs in dynein RNAi cells, revealing activation of an M-phase specific mechanism of ER-MT association. ER redistribution to spindle poles also did not require non-claret disjunctional (ncd), the other known Drosophila MT minus-end motor, nor Klp61F, a MT plus-end motor that generates spindle poleward forces. Collectively, our results suggest that a novel, M-phase specific mechanism of ER-MT association that is independent of MT minus-end motors is required for proper ER partitioning in dividing cells.
Collapse
Affiliation(s)
- Darya Karabasheva
- Department of Anatomy, Physiology, and Genetics, Uniformed Services University of the Health Sciences, F. Edward Hébert School of Medicine, Bethesda, MD, 20814, USA
| | - Jeremy T Smyth
- Department of Anatomy, Physiology, and Genetics, Uniformed Services University of the Health Sciences, F. Edward Hébert School of Medicine, Bethesda, MD, 20814, USA.
| |
Collapse
|
40
|
Saraste J, Prydz K. A New Look at the Functional Organization of the Golgi Ribbon. Front Cell Dev Biol 2019; 7:171. [PMID: 31497600 PMCID: PMC6713163 DOI: 10.3389/fcell.2019.00171] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Accepted: 08/07/2019] [Indexed: 12/14/2022] Open
Abstract
A characteristic feature of vertebrate cells is a Golgi ribbon consisting of multiple cisternal stacks connected into a single-copy organelle next to the centrosome. Despite numerous studies, the mechanisms that link the stacks together and the functional significance of ribbon formation remain poorly understood. Nevertheless, these questions are of considerable interest, since there is increasing evidence that Golgi fragmentation – the unlinking of the stacks in the ribbon – is intimately connected not only to normal physiological processes, such as cell division and migration, but also to pathological states, including neurodegeneration and cancer. Challenging a commonly held view that ribbon architecture involves the formation of homotypic tubular bridges between the Golgi stacks, we present an alternative model, based on direct interaction between the biosynthetic (pre-Golgi) and endocytic (post-Golgi) membrane networks and their connection with the centrosome. We propose that the central domains of these permanent pre- and post-Golgi networks function together in the biogenesis and maintenance of the more transient Golgi stacks, and thereby establish “linker compartments” that dynamically join the stacks together. This model provides insight into the reversible fragmentation of the Golgi ribbon that takes place in dividing and migrating cells and its regulation along a cell surface – Golgi – centrosome axis. Moreover, it helps to understand transport pathways that either traverse or bypass the Golgi stacks and the positioning of the Golgi apparatus in differentiated neuronal, epithelial, and muscle cells.
Collapse
Affiliation(s)
- Jaakko Saraste
- Department of Biomedicine and Molecular Imaging Center, University of Bergen, Bergen, Norway
| | - Kristian Prydz
- Department of Biosciences, University of Oslo, Oslo, Norway
| |
Collapse
|
41
|
Mascanzoni F, Ayala I, Colanzi A. Organelle Inheritance Control of Mitotic Entry and Progression: Implications for Tissue Homeostasis and Disease. Front Cell Dev Biol 2019; 7:133. [PMID: 31396510 PMCID: PMC6664238 DOI: 10.3389/fcell.2019.00133] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Accepted: 07/04/2019] [Indexed: 12/12/2022] Open
Abstract
The Golgi complex (GC), in addition to its well-known role in membrane traffic, is also actively involved in the regulation of mitotic entry and progression. In particular, during the G2 phase of the cell cycle, the Golgi ribbon is unlinked into isolated stacks. Importantly, this ribbon cleavage is required for G2/M transition, indicating that a "Golgi mitotic checkpoint" controls the correct segregation of this organelle. Then, during mitosis, the isolated Golgi stacks are disassembled, and this process is required for spindle formation. Moreover, recent evidence indicates that also proper mitotic segregation of other organelles, such as mitochondria, endosomes, and peroxisomes, is required for correct mitotic progression and/or spindle formation. Collectively, these observations imply that in addition to the control of chromosomes segregation, which is required to preserve the genetic information, the cells actively monitor the disassembly and redistribution of subcellular organelles in mitosis. Here, we provide an overview of the major structural reorganization of the GC and other organelles during G2/M transition and of their regulatory mechanisms, focusing on novel findings that have shed light on the basic processes that link organelle inheritance to mitotic progression and spindle formation, and discussing their implications for tissue homeostasis and diseases.
Collapse
Affiliation(s)
| | | | - Antonino Colanzi
- Institute of Biochemistry and Cell Biology, National Research Council, Naples, Italy
| |
Collapse
|
42
|
Keller M, Dubois F, Teulier S, Martin APJ, Levallet J, Maille E, Brosseau S, Elie N, Hergovich A, Bergot E, Camonis J, Zalcman G, Levallet G. NDR2 kinase contributes to cell invasion and cytokinesis defects induced by the inactivation of RASSF1A tumor-suppressor gene in lung cancer cells. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:158. [PMID: 30979377 PMCID: PMC6461807 DOI: 10.1186/s13046-019-1145-8] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Accepted: 03/19/2019] [Indexed: 12/14/2022]
Abstract
Background RASSF1A, a tumor suppressor gene, is frequently inactivated in lung cancer leading to a YAP-dependent epithelial-mesenchymal transition (EMT). Such effects are partly due to the inactivation of the anti-migratory RhoB GTPase via the inhibitory phosphorylation of GEF-H1, the GDP/GTP exchange factor for RhoB. However, the kinase responsible for RhoB/GEF-H1 inactivation in RASSF1A-depleted cells remained unknown. Methods NDR1/2 inactivation by siRNA or shRNA effects on epithelial-mesenchymal transition, invasion, xenograft formation and growth in SCID−/− Beige mice, apoptosis, proliferation, cytokinesis, YAP/TAZ activation were investigated upon RASSF1A loss in human bronchial epithelial cells (HBEC). Results We demonstrate here that depletion of the YAP-kinases NDR1/2 reverts migration and metastatic properties upon RASSF1A loss in HBEC. We show that NDR2 interacts directly with GEF-H1 (which contains the NDR phosphorylation consensus motif HXRXXS/T), leading to GEF-H1 phosphorylation. We further report that the RASSF1A/NDR2/GEF-H1/RhoB/YAP axis is involved in proper cytokinesis in human bronchial cells, since chromosome proper segregation are NDR-dependent upon RASSF1A or GEF-H1 loss in HBEC. Conclusion To summarize, our data support a model in which, upon RASSF1A silencing, NDR2 gets activated, phosphorylates and inactivates GEF-H1, leading to RhoB inactivation. This cascade induced by RASSF1A loss in bronchial cells is responsible for metastasis properties, YAP activation and cytokinesis defects. Electronic supplementary material The online version of this article (10.1186/s13046-019-1145-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Maureen Keller
- Normandie University, UNICAEN, UMR 1086 INSERM, F-14032, Caen, France.,Normandie University, UNICAEN, UPRES-EA-2608, F-14032, Caen, France
| | - Fatéméh Dubois
- Normandie University, UNICAEN, CEA, CNRS, ISTCT/CERVOxy group, GIP CYCERON, Avenue H.Becquerel- 14074, F-14000, Caen, France.,Service d'Anatomie et Cytologie Pathologique, CHU de Caen, F-14033, Caen, France
| | - Sylvain Teulier
- Normandie University, UNICAEN, CEA, CNRS, ISTCT/CERVOxy group, GIP CYCERON, Avenue H.Becquerel- 14074, F-14000, Caen, France
| | - Alexandre P J Martin
- U830 INSERM, "Génétique et Biologie des cancers" Centre de Recherche, Institut Curie, Paris, France
| | - Jérôme Levallet
- Normandie University, UNICAEN, UPRES-EA-2608, F-14032, Caen, France
| | - Elodie Maille
- Normandie University, UNICAEN, UMR 1086 INSERM, F-14032, Caen, France.,Normandie University, UNICAEN, UPRES-EA-2608, F-14032, Caen, France
| | - Solenn Brosseau
- Normandie University, UNICAEN, UMR 1086 INSERM, F-14032, Caen, France.,Normandie University, UNICAEN, UPRES-EA-2608, F-14032, Caen, France.,Service d'oncologie thoracique, CIC 1425, Hôpital Bichat-Claude Bernard, AP-HP, Université Paris-Diderot, Paris, France
| | - Nicolas Elie
- Normandie Univ, UNICAEN, SFR ICORE, Plateau CMABio3, F-14032, Caen, France
| | | | - Emmanuel Bergot
- Normandie University, UNICAEN, CEA, CNRS, ISTCT/CERVOxy group, GIP CYCERON, Avenue H.Becquerel- 14074, F-14000, Caen, France.,Service de Pneumologie-Oncologie thoracique, CHU de Caen, F-14033, Caen, France
| | - Jacques Camonis
- U830 INSERM, "Génétique et Biologie des cancers" Centre de Recherche, Institut Curie, Paris, France
| | - Gérard Zalcman
- U830 INSERM, "Génétique et Biologie des cancers" Centre de Recherche, Institut Curie, Paris, France.,Service d'oncologie thoracique, CIC 1425, Hôpital Bichat-Claude Bernard, AP-HP, Université Paris-Diderot, Paris, France
| | - Guénaëlle Levallet
- Normandie University, UNICAEN, CEA, CNRS, ISTCT/CERVOxy group, GIP CYCERON, Avenue H.Becquerel- 14074, F-14000, Caen, France. .,Service d'Anatomie et Cytologie Pathologique, CHU de Caen, F-14033, Caen, France.
| |
Collapse
|
43
|
Chumová J, Kourová H, Trögelová L, Halada P, Binarová P. Microtubular and Nuclear Functions of γ-Tubulin: Are They LINCed? Cells 2019; 8:cells8030259. [PMID: 30893853 PMCID: PMC6468392 DOI: 10.3390/cells8030259] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Revised: 03/07/2019] [Accepted: 03/14/2019] [Indexed: 01/02/2023] Open
Abstract
γ-Tubulin is a conserved member of the tubulin superfamily with a function in microtubule nucleation. Proteins of γ-tubulin complexes serve as nucleation templates as well as a majority of other proteins contributing to centrosomal and non-centrosomal nucleation, conserved across eukaryotes. There is a growing amount of evidence of γ-tubulin functions besides microtubule nucleation in transcription, DNA damage response, chromatin remodeling, and on its interactions with tumor suppressors. However, the molecular mechanisms are not well understood. Furthermore, interactions with lamin and SUN proteins of the LINC complex suggest the role of γ-tubulin in the coupling of nuclear organization with cytoskeletons. γ-Tubulin that belongs to the clade of eukaryotic tubulins shows characteristics of both prokaryotic and eukaryotic tubulins. Both human and plant γ-tubulins preserve the ability of prokaryotic tubulins to assemble filaments and higher-order fibrillar networks. γ-Tubulin filaments, with bundling and aggregating capacity, are suggested to perform complex scaffolding and sequestration functions. In this review, we discuss a plethora of γ-tubulin molecular interactions and cellular functions, as well as recent advances in understanding the molecular mechanisms behind them.
Collapse
Affiliation(s)
- Jana Chumová
- Institute of Microbiology of the Czech Academy of Sciences, Vídeňská 1083, 142 20 Prague, Czech Republic.
| | - Hana Kourová
- Institute of Microbiology of the Czech Academy of Sciences, Vídeňská 1083, 142 20 Prague, Czech Republic.
| | - Lucie Trögelová
- Institute of Microbiology of the Czech Academy of Sciences, Vídeňská 1083, 142 20 Prague, Czech Republic.
| | - Petr Halada
- Institute of Microbiology of the Czech Academy of Sciences, Vídeňská 1083, 142 20 Prague, Czech Republic.
| | - Pavla Binarová
- Institute of Microbiology of the Czech Academy of Sciences, Vídeňská 1083, 142 20 Prague, Czech Republic.
| |
Collapse
|
44
|
Rosselló CA, Lindström L, Eklund G, Corvaisier M, Kristensson MA. γ-Tubulin⁻γ-Tubulin Interactions as the Basis for the Formation of a Meshwork. Int J Mol Sci 2018; 19:ijms19103245. [PMID: 30347727 PMCID: PMC6214090 DOI: 10.3390/ijms19103245] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Revised: 10/02/2018] [Accepted: 10/16/2018] [Indexed: 12/14/2022] Open
Abstract
In cytoplasm, protein γ-tubulin joins with various γ-tubulin complex proteins (GCPs) to form a heterotetramer γ-tubulin small complex (γ-TuSC) that can grow into a ring-shaped structure called the γ-tubulin ring complex (γ-TuRC). Both γ-TuSC and γ-TuRC are required for microtubule nucleation. Recent knowledge on γ-tubulin with regard to its cellular functions beyond participation in its creation of microtubules suggests that this protein forms a cellular meshwork. The present review summarizes the recognized functions of γ-tubulin and aims to unite the current views on this protein.
Collapse
Affiliation(s)
- Catalina Ana Rosselló
- Molecular Pathology, Department of Translational Medicine, Lund University, Skåne University Hospital, 20502 Malmö, Sweden.
| | - Lisa Lindström
- Molecular Pathology, Department of Translational Medicine, Lund University, Skåne University Hospital, 20502 Malmö, Sweden.
| | - Greta Eklund
- Molecular Pathology, Department of Translational Medicine, Lund University, Skåne University Hospital, 20502 Malmö, Sweden.
| | - Matthieu Corvaisier
- Molecular Pathology, Department of Translational Medicine, Lund University, Skåne University Hospital, 20502 Malmö, Sweden.
| | - Maria Alvarado Kristensson
- Molecular Pathology, Department of Translational Medicine, Lund University, Skåne University Hospital, 20502 Malmö, Sweden.
| |
Collapse
|
45
|
Alvarado-Kristensson M. γ-tubulin as a signal-transducing molecule and meshwork with therapeutic potential. Signal Transduct Target Ther 2018; 3:24. [PMID: 30221013 PMCID: PMC6137058 DOI: 10.1038/s41392-018-0021-x] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Revised: 04/23/2018] [Accepted: 05/06/2018] [Indexed: 01/05/2023] Open
Abstract
Knowledge of γ-tubulin is increasing with regard to the cellular functions of this protein beyond its participation in microtubule nucleation. γ-Tubulin expression is altered in various malignancies, and changes in the TUBG1 gene have been found in patients suffering from brain malformations. This review recapitulates the known functions of γ-tubulin in cellular homeostasis and discusses the possible influence of the protein on disease development and cancer.
Collapse
Affiliation(s)
- Maria Alvarado-Kristensson
- Molecular Pathology, Department of Translational Medicine, Lund University, Skåne University Hospital, Malmö, 20502 Sweden
| |
Collapse
|
46
|
Xie S, Reinecke JB, Farmer T, Bahl K, Yeow I, Nichols BJ, McLamarrah TA, Naslavsky N, Rogers GC, Caplan S. Vesicular trafficking plays a role in centriole disengagement and duplication. Mol Biol Cell 2018; 29:2622-2631. [PMID: 30188792 PMCID: PMC6249839 DOI: 10.1091/mbc.e18-04-0241] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Centrosomes are the major microtubule-nucleating and microtubule-organizing centers of cells and play crucial roles in microtubule anchoring, organelle positioning, and ciliogenesis. At the centrosome core lies a tightly associated or “engaged” mother–daughter centriole pair. During mitotic exit, removal of centrosomal proteins pericentrin and Cep215 promotes “disengagement” by the dissolution of intercentriolar linkers, ensuring a single centriole duplication event per cell cycle. Herein, we explore a new mechanism involving vesicular trafficking for the removal of centrosomal Cep215. Using small interfering RNA and CRISPR/Cas9 gene-edited cells, we show that the endocytic protein EHD1 regulates Cep215 transport from centrosomes to the spindle midbody, thus facilitating disengagement and duplication. We demonstrate that EHD1 and Cep215 interact and show that Cep215 displays increased localization to vesicles containing EHD1 during mitosis. Moreover, Cep215-containing vesicles are positive for internalized transferrin, demonstrating their endocytic origin. Thus, we describe a novel relationship between endocytic trafficking and the centrosome cycle, whereby vesicles of endocytic origin are used to remove key regulatory proteins from centrosomes to control centriole duplication.
Collapse
Affiliation(s)
- Shuwei Xie
- Department of Biochemistry and Molecular Biology and Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198-5870
| | - James B Reinecke
- Department of Biochemistry and Molecular Biology and Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198-5870
| | - Trey Farmer
- Department of Biochemistry and Molecular Biology and Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198-5870
| | - Kriti Bahl
- Department of Biochemistry and Molecular Biology and Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198-5870
| | - Ivana Yeow
- MRC-Laboratory of Molecular Biology, Cambridge CB2 2QH, United Kingdom
| | | | - Tiffany A McLamarrah
- Department of Cellular and Molecular Medicine, University of Arizona Cancer Center, University of Arizona, Tucson, AZ 85724
| | - Naava Naslavsky
- Department of Biochemistry and Molecular Biology and Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198-5870
| | - Gregory C Rogers
- Department of Cellular and Molecular Medicine, University of Arizona Cancer Center, University of Arizona, Tucson, AZ 85724
| | - Steve Caplan
- Department of Biochemistry and Molecular Biology and Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198-5870
| |
Collapse
|
47
|
Biocompatible PEGylated Gold nanorods function As cytokinesis inhibitors to suppress angiogenesis. Biomaterials 2018; 178:23-35. [DOI: 10.1016/j.biomaterials.2018.06.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Revised: 05/16/2018] [Accepted: 06/06/2018] [Indexed: 12/17/2022]
|
48
|
Kinesin-2 Controls the Motility of RAB5 Endosomes and Their Association with the Spindle in Mitosis. Int J Mol Sci 2018; 19:ijms19092575. [PMID: 30200238 PMCID: PMC6163544 DOI: 10.3390/ijms19092575] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Revised: 08/08/2018] [Accepted: 08/27/2018] [Indexed: 12/19/2022] Open
Abstract
RAB5 is a small GTPase that belongs to the wide family of Rab proteins and localizes on early endosomes. In its active GTP-bound form, RAB5 recruits downstream effectors that, in turn, are responsible for distinct aspects of early endosome function, including their movement along microtubules. We previously reported that, at the onset of mitosis, RAB5positive vesicles cluster around the spindle poles and, during metaphase, move along spindle microtubules. RNAi-mediated depletion of the three RAB5 isoforms delays nuclear envelope breakdown at prophase and severely affects chromosome alignment and segregation. Here we show that depletion of the Kinesin-2 motor complex impairs long-range movement of RAB5 endosomes in interphase cells and prevents localization of these vesicles at the spindle during metaphase. Similarly to the effect caused by RAB5 depletion, functional ablation of Kinesin-2 delays nuclear envelope breakdown resulting in prolonged prophase. Altogether these findings suggest that endosomal transport at the onset of mitosis is required to control timing of nuclear envelope breakdown.
Collapse
|
49
|
Hua K, Ferland RJ. Primary Cilia Reconsidered in the Context of Ciliopathies: Extraciliary and Ciliary Functions of Cilia Proteins Converge on a Polarity theme? Bioessays 2018; 40:e1700132. [PMID: 29882973 PMCID: PMC6239423 DOI: 10.1002/bies.201700132] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Revised: 05/09/2018] [Indexed: 12/13/2022]
Abstract
Once dismissed as vestigial organelles, primary cilia have garnered the interest of scientists, given their importance in development/signaling, and for their implication in a new disease category known as ciliopathies. However, many, if not all, "cilia" proteins also have locations/functions outside of the primary cilium. These extraciliary functions can complicate the interpretation of a particular ciliopathy phenotype: it may be a result of defects at the cilium and/or at extraciliary locations, and it could be broadly related to a unifying cellular process for these proteins, such as polarity. Assembly of a cilium has many similarities to the development of other polarized structures. This evolutionarily preserved process for the assembly of polarized cell structures offers a perspective on how the cilium may have evolved. We hypothesize that cilia proteins are critical for cell polarity, and that core polarity proteins may have been specialized to form various cellular protrusions, including primary cilia.
Collapse
Affiliation(s)
- Kiet Hua
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, Albany, New York, USA, 12208
| | - Russell J Ferland
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, Albany, New York, USA, 12208
- Department of Neurology, Albany Medical College, Albany, New York, USA, 12208
| |
Collapse
|
50
|
Gong X, Liu J, Zhang X, Dong F, Liu Y, Wang P. Rab11 Functions as an Oncoprotein via Nuclear Factor kappa B (NF-κB) Signaling Pathway in Human Bladder Carcinoma. Med Sci Monit 2018; 24:5093-5101. [PMID: 30032159 PMCID: PMC6067026 DOI: 10.12659/msm.911454] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Background Elevated expression of Rab11 has been reported in different human cancers, including human bladder carcinoma. This study, we investigated the biological effects and mechanism of Rab11 overexpression in human bladder carcinoma for the first time. Material/Methods Rab11 expression in bladder cancer tissues was detected using immunohistochemistry and Western blot analysis. Then, Rab11 expression was inhibited in T24 cells and it was overexpressed in BIU-87 cells. The effects of Rab11 perturbations on cell growth rate and invasion were analyzed by CCK8, cell cycle assay, and matrix gel invasion assay. MMP-9, cyclin E, and cyclin D1 levels were studied using Western blot and qPCR. NF-κB activity was studied by luciferase assay. Results High expression of Rab11 was detected in 41.5% (66/159) of tumor specimens. We found a significant correlation between high Rab11 expression and depth of tumor invasion (P=0.004). Rab11 overexpression was observed to promote the growth rate and invasiveness of cancer cells through upregulation of MMP9, cyclin E, and cyclin D1 levels. Rab11 overexpression further elevated NF-κB reporter activity and enhanced p-IκB expression. Use of BAY 11-7082, a noted NF-κB inhibitor, partially abolished overexpression of MMP9 and cyclin D1 by Rab11. Conclusions Our research proved that high Rab11 expression enhances cellular multiplication and invasiveness of bladder cancer, possibly by regulating the NF-κB signaling pathway.
Collapse
Affiliation(s)
- Xue Gong
- Department of Urology, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning, China (mainland)
| | - Jia Liu
- Department of Urology, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning, China (mainland)
| | - Xiling Zhang
- Department of Urology, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning, China (mainland)
| | - Fengming Dong
- Department of Urology, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning, China (mainland)
| | - Yili Liu
- Department of Urology, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning, China (mainland)
| | - Ping Wang
- Department of Urology, The Fourth Affiliated Hospital of China Medical University, Shenyang, China (mainland)
| |
Collapse
|