201
|
Sala R, Farrell KC, Stearns T. Growth disadvantage associated with centrosome amplification drives population-level centriole number homeostasis. Mol Biol Cell 2020; 31:2646-2656. [PMID: 32966175 PMCID: PMC7927180 DOI: 10.1091/mbc.e19-04-0195] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The centriole duplication cycle normally ensures that centriole number is maintained at two centrioles per G1 cell. However, some circumstances can result in an aberrant increase in centriole number—a phenotype that is particularly prevalent in several types of cancer. Following an artificial increase in centriole number without tetraploidization due to transient overexpression of the kinase PLK4, human cells return to a normal centriole number during the proliferation of the population. We examine the mechanisms responsible for this return to normal centriole number at the population level in human retinal pigment epithelial cells. We find that the return to normal centriole number in the population of induced cells cannot be explained by limited duplication of centrioles, instability of extra centrioles, or by grossly asymmetric segregation of extra centrioles in mitosis. However, cells with extra centrioles display heterogenous phenotypes including extended cell cycle arrest, longer interphase durations, and death, which overall results in a proliferative disadvantage relative to normal cells in the population. Although about half of cells with extra centrioles in a population were able to divide, the extent of the disadvantages conferred by other fates is sufficient to account for the observed rate of return to normal centriole number. These results suggest that only under conditions of positive selection for cells with extra centrioles, continuous generation of such centrioles, or alleviation of the disadvantageous growth phenotypes would they be maintained in a population.
Collapse
Affiliation(s)
- Roberta Sala
- Department of Biology, Stanford University, Stanford, CA 94305
| | - K C Farrell
- Department of Biology, Stanford University, Stanford, CA 94305
| | - Tim Stearns
- Department of Biology, Stanford University, Stanford, CA 94305.,Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305
| |
Collapse
|
202
|
Halder P, Khatun S, Majumder S. Freeing the brake: Proliferation needs primary cilium to disassemble. J Biosci 2020. [DOI: 10.1007/s12038-020-00090-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
203
|
Yamamoto S, Kitagawa D. Emerging insights into symmetry breaking in centriole duplication: updated view on centriole duplication theory. Curr Opin Struct Biol 2020; 66:8-14. [PMID: 32956908 DOI: 10.1016/j.sbi.2020.08.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Revised: 08/22/2020] [Accepted: 08/25/2020] [Indexed: 11/15/2022]
Abstract
Centriole duplication occurs once per cell cycle. Since only a single daughter centriole is assembled adjacent to each mother centriole, symmetry around the mother centriole must be broken in the process of centriole duplication. Recent studies have established that Plk4, a master kinase for centriole duplication, can self-assemble into condensates, and have suggested that this Plk4 self-assembly is the key to symmetry breaking. Here, we present the current hypotheses for how Plk4 could break symmetry around the mother centriole via autonomous regulation. After this initial symmetry-breaking process, the ring-to-dot conversion of Plk4 around the mother centriole completes the selection of the site for procentriole formation. We also discuss how this dynamic transition contributes to the strict regulation of centriole duplication.
Collapse
Affiliation(s)
- Shohei Yamamoto
- Department of Physiological Chemistry, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Hongo, Tokyo 113-0033, Japan
| | - Daiju Kitagawa
- Department of Physiological Chemistry, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Hongo, Tokyo 113-0033, Japan.
| |
Collapse
|
204
|
Vakonakis I. The centriolar cartwheel structure: symmetric, stacked, and polarized. Curr Opin Struct Biol 2020; 66:1-7. [PMID: 32956907 DOI: 10.1016/j.sbi.2020.08.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 08/24/2020] [Accepted: 08/25/2020] [Indexed: 11/29/2022]
Abstract
An accurate centriolar structure is crucial for organelle function, necessitating the existence of molecular mechanisms for the tight control of centriole assembly. Formation of an initial scaffold, the cartwheel, assists the correct placement of centriolar proteins during assembly and templates key structural parameters of the organelle. Past work illustrated how cartwheel and centriolar symmetry are linked, and grounded organelle symmetry and diameter to the properties of the centriolar protein SAS-6. However, questions remained over how centriole polarity and length are controlled. Recent advances in resolving cartwheel structure and cell biology showed that these assemblies are polarized and that their length is under the control of a homeostatic mechanism. These cartwheel properties may, in turn, influence the centriolar polarity and length.
Collapse
Affiliation(s)
- Ioannis Vakonakis
- Department of Biochemistry, University of Oxford, Oxford OX1 3QU, United Kingdom.
| |
Collapse
|
205
|
Steib E, Laporte MH, Gambarotto D, Olieric N, Zheng C, Borgers S, Olieric V, Le Guennec M, Koll F, Tassin AM, Steinmetz MO, Guichard P, Hamel V. WDR90 is a centriolar microtubule wall protein important for centriole architecture integrity. eLife 2020; 9:57205. [PMID: 32946374 PMCID: PMC7500955 DOI: 10.7554/elife.57205] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 08/22/2020] [Indexed: 12/13/2022] Open
Abstract
Centrioles are characterized by a nine-fold arrangement of microtubule triplets held together by an inner protein scaffold. These structurally robust organelles experience strenuous cellular processes such as cell division or ciliary beating while performing their function. However, the molecular mechanisms underlying the stability of microtubule triplets, as well as centriole architectural integrity remain poorly understood. Here, using ultrastructure expansion microscopy for nanoscale protein mapping, we reveal that POC16 and its human homolog WDR90 are components of the microtubule wall along the central core region of the centriole. We further found that WDR90 is an evolutionary microtubule associated protein. Finally, we demonstrate that WDR90 depletion impairs the localization of inner scaffold components, leading to centriole structural abnormalities in human cells. Altogether, this work highlights that WDR90 is an evolutionary conserved molecular player participating in centriole architecture integrity. Cells are made up of compartments called organelles that perform specific roles. A cylindrical organelle called the centriole is important for a number of cellular processes, ranging from cell division to movement and signaling. Each centriole contains nine blades made up of protein filaments called microtubules, which link together to form a cylinder. This well-known structure can be found in a variety of different species. Yet, it is unclear how centrioles are able to maintain this stable architecture whilst carrying out their various different cell roles. In early 2020, a group of researchers discovered a scaffold protein at the center of centrioles that helps keep the microtubule blades stable. Further investigation suggested that another protein called WDR90 may also help centrioles sustain their cylindrical shape. However, the exact role of this protein was poorly understood. To determine the role of WDR90, Steib et al. – including many of the researchers involved in the 2020 study – used a method called Ultrastructure Expansion Microscopy to precisely locate the WDR90 protein in centrioles. This revealed that WDR90 is located on the microtubule wall of centrioles in green algae and human cells grown in the lab. Further experiments showed that the protein binds directly to microtubules and that removing WDR90 from human cells causes centrioles to lose their scaffold proteins and develop structural defects. This investigation provides fundamental insights into the structure and stability of centrioles. It shows that single proteins are key components in supporting the structural integrity of organelles and shaping their overall architecture. Furthermore, these findings demonstrate how ultrastructure expansion microscopy can be used to determine the role of individual proteins within a complex structure.
Collapse
Affiliation(s)
- Emmanuelle Steib
- University of Geneva, Department of Cell Biology, Sciences III, Geneva, Switzerland
| | - Marine H Laporte
- University of Geneva, Department of Cell Biology, Sciences III, Geneva, Switzerland
| | - Davide Gambarotto
- University of Geneva, Department of Cell Biology, Sciences III, Geneva, Switzerland
| | - Natacha Olieric
- Laboratory of Biomolecular Research, Division of Biology and Chemistry, Paul Scherrer Institut, Villigen, Switzerland
| | - Celine Zheng
- Laboratory of Biomolecular Research, Division of Biology and Chemistry, Paul Scherrer Institut, Villigen, Switzerland
| | - Susanne Borgers
- University of Geneva, Department of Cell Biology, Sciences III, Geneva, Switzerland
| | - Vincent Olieric
- Swiss Light Source, Paul Scherrer Institut, Villigen, Switzerland
| | - Maeva Le Guennec
- University of Geneva, Department of Cell Biology, Sciences III, Geneva, Switzerland
| | - France Koll
- Institute for Integrative Biology of the Cell (I2BC), CEA, CNRS, Univ. Paris Sud, Université Paris-Saclay, Gif sur Yvette, France
| | - Anne-Marie Tassin
- Institute for Integrative Biology of the Cell (I2BC), CEA, CNRS, Univ. Paris Sud, Université Paris-Saclay, Gif sur Yvette, France
| | - Michel O Steinmetz
- Laboratory of Biomolecular Research, Division of Biology and Chemistry, Paul Scherrer Institut, Villigen, Switzerland.,Biozentrum, University of Basel, Basel, Switzerland
| | - Paul Guichard
- University of Geneva, Department of Cell Biology, Sciences III, Geneva, Switzerland
| | - Virginie Hamel
- University of Geneva, Department of Cell Biology, Sciences III, Geneva, Switzerland
| |
Collapse
|
206
|
Wang T, Zou Y, Huang N, Teng J, Chen J. CCDC84 Acetylation Oscillation Regulates Centrosome Duplication by Modulating HsSAS-6 Degradation. Cell Rep 2020; 29:2078-2091.e5. [PMID: 31722219 DOI: 10.1016/j.celrep.2019.10.028] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 09/16/2019] [Accepted: 10/07/2019] [Indexed: 01/14/2023] Open
Abstract
In animal cells, centriole number is strictly controlled in order to guarantee faithful cell division and genetic stability, but the mechanism by which the accuracy of centrosome duplication is maintained is not fully understood. Here, we show that CCDC84 constrains centriole number by modulating APC/CCdh1-mediated HsSAS-6 degradation. More importantly, CCDC84 acetylation oscillates throughout the cell cycle, and the acetylation state of CCDC84 at lysine 31 is regulated by the deacetylase SIRT1 and the acetyltransferase NAT10. Deacetylated CCDC84 is responsible for its centrosome targeting, and acetylated CCDC84 promotes HsSAS-6 ubiquitination by enhancing the binding affinity of HsSAS-6 for Cdh1. Our findings shed new light on the function of (de)acetylation in centriole number regulation as well as refine the established centrosome duplication model.
Collapse
Affiliation(s)
- Tianning Wang
- Key Laboratory of Cell Proliferation and Differentiation of the Ministry of Education and State Key Laboratory of Membrane Biology, College of Life Sciences, Peking University, Beijing 100871, China
| | - Yuhong Zou
- Key Laboratory of Cell Proliferation and Differentiation of the Ministry of Education and State Key Laboratory of Membrane Biology, College of Life Sciences, Peking University, Beijing 100871, China
| | - Ning Huang
- Key Laboratory of Cell Proliferation and Differentiation of the Ministry of Education and State Key Laboratory of Membrane Biology, College of Life Sciences, Peking University, Beijing 100871, China
| | - Junlin Teng
- Key Laboratory of Cell Proliferation and Differentiation of the Ministry of Education and State Key Laboratory of Membrane Biology, College of Life Sciences, Peking University, Beijing 100871, China.
| | - Jianguo Chen
- Key Laboratory of Cell Proliferation and Differentiation of the Ministry of Education and State Key Laboratory of Membrane Biology, College of Life Sciences, Peking University, Beijing 100871, China; Center for Quantitative Biology, Peking University, Beijing 100871, China.
| |
Collapse
|
207
|
McLamarrah TA, Speed SK, Ryniawec JM, Buster DW, Fagerstrom CJ, Galletta BJ, Rusan NM, Rogers GC. A molecular mechanism for the procentriole recruitment of Ana2. J Cell Biol 2020; 219:132764. [PMID: 31841145 PMCID: PMC7041687 DOI: 10.1083/jcb.201905172] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Revised: 10/24/2019] [Accepted: 11/12/2019] [Indexed: 12/15/2022] Open
Abstract
McLamarrah et al. characterize an early step in centriole duplication. They show that Plk4 hyperphosphorylates Ana2, which increases the affinity of Ana2 for the G-box domain of Sas4, promoting Ana2’s accumulation at the procentriole and, consequently, daughter centriole formation. During centriole duplication, a preprocentriole forms at a single site on the mother centriole through a process that includes the hierarchical recruitment of a conserved set of proteins, including the Polo-like kinase 4 (Plk4), Ana2/STIL, and the cartwheel protein Sas6. Ana2/STIL is critical for procentriole assembly, and its recruitment is controlled by the kinase activity of Plk4, but how this works remains poorly understood. A structural motif called the G-box in the centriole outer wall protein Sas4 interacts with a short region in the N terminus of Ana2/STIL. Here, we show that binding of Ana2 to the Sas4 G-box enables hyperphosphorylation of the Ana2 N terminus by Plk4. Hyperphosphorylation increases the affinity of the Ana2–G-box interaction, and, consequently, promotes the accumulation of Ana2 at the procentriole to induce daughter centriole formation.
Collapse
Affiliation(s)
- Tiffany A McLamarrah
- Department of Cellular and Molecular Medicine, University of Arizona Cancer Center, University of Arizona, Tucson, AZ
| | - Sarah K Speed
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| | - John M Ryniawec
- Department of Cellular and Molecular Medicine, University of Arizona Cancer Center, University of Arizona, Tucson, AZ
| | - Daniel W Buster
- Department of Cellular and Molecular Medicine, University of Arizona Cancer Center, University of Arizona, Tucson, AZ
| | - Carey J Fagerstrom
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Brian J Galletta
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Nasser M Rusan
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Gregory C Rogers
- Department of Cellular and Molecular Medicine, University of Arizona Cancer Center, University of Arizona, Tucson, AZ
| |
Collapse
|
208
|
TRIM37 controls cancer-specific vulnerability to PLK4 inhibition. Nature 2020; 585:440-446. [PMID: 32908304 PMCID: PMC7501188 DOI: 10.1038/s41586-020-2710-1] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Accepted: 06/09/2020] [Indexed: 12/21/2022]
Abstract
Centrosomes catalyze microtubule formation for mitotic spindle assembly1. Centrosomes duplicate once per cell cycle in a process controlled the kinase PLK42,3. Following chemical PLK4 inhibition, cell division in the absence of centrosome duplication generates centrosome-less cells that exhibit delayed, acentrosomal spindle assembly4. Whether PLK4 inhibitors can be leveraged for cancer treatment is not yet clear. Here, we show that acentrosomal spindle assembly following PLK4 inhibition depends on levels of the centrosomal ubiquitin ligase TRIM37. Low TRIM37 accelerates acentrosomal spindle assembly and improves proliferation following PLK4 inhibition, whereas high TRIM37 inhibits acentrosomal spindle assembly, leading to mitotic failure and cessation of proliferation. The Chr17q region containing the TRIM37 gene is frequently amplified in neuroblastoma and in breast cancer5–8, which renders these cancer types highly sensitive to PLK4 inhibition. TRIM37 inactivation improves acentrosomal mitosis because TRIM37 prevents PLK4 self-assembly into centrosome-independent condensates that serve as ectopic microtubule-organizing centers. By contrast, elevated TRIM37 expression inhibits acentrosomal spindle assembly via a distinct mechanism that involves degradation of the centrosomal component CEP192. Thus, TRIM37 is a critical determinant of mitotic vulnerability to PLK4 inhibition. Linkage of TRIM37 to prevalent cancer-associated genomic changes, including 17q gain in neuroblastoma and 17q23 amplification in breast cancer, may offer an opportunity to use PLK4 inhibition to trigger selective mitotic failure and provide new avenues to treatments for these cancers.
Collapse
|
209
|
Ching K, Stearns T. Centrioles are amplified in cycling progenitors of olfactory sensory neurons. PLoS Biol 2020; 18:e3000852. [PMID: 32931487 PMCID: PMC7518617 DOI: 10.1371/journal.pbio.3000852] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 09/25/2020] [Accepted: 08/20/2020] [Indexed: 11/19/2022] Open
Abstract
Olfaction in most animals is mediated by neurons bearing cilia that are accessible to the environment. Olfactory sensory neurons (OSNs) in chordates usually have multiple cilia, each with a centriole at its base. OSNs differentiate from stem cells in the olfactory epithelium, and how the epithelium generates cells with many centrioles is not yet understood. We show that centrioles are amplified via centriole rosette formation in both embryonic development and turnover of the olfactory epithelium in adult mice, and rosette-bearing cells often have free centrioles in addition. Cells with amplified centrioles can go on to divide, with centrioles clustered at each pole. Additionally, we found that centrioles are amplified in immediate neuronal precursors (INPs) concomitant with elevation of mRNA for polo-like kinase 4 (Plk4) and SCL/Tal1-interrupting locus gene (Stil), key regulators of centriole duplication. These results support a model in which centriole amplification occurs during a transient state characterized by elevated Plk4 and Stil in early INP cells. These cells then go on to divide at least once to become OSNs, demonstrating that cell division with amplified centrioles, known to be tolerated in disease states, can occur as part of a normal developmental program.
Collapse
Affiliation(s)
- Kaitlin Ching
- Department of Biology, Stanford University, Stanford, California, United States of America
| | - Tim Stearns
- Department of Biology, Stanford University, Stanford, California, United States of America
- Department of Genetics, Stanford University School of Medicine, Stanford, California, United States of America
| |
Collapse
|
210
|
Goryachev AB, Leda M. Compete or Coexist? Why the Same Mechanisms of Symmetry Breaking Can Yield Distinct Outcomes. Cells 2020; 9:E2011. [PMID: 32882972 PMCID: PMC7563139 DOI: 10.3390/cells9092011] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 08/27/2020] [Accepted: 08/28/2020] [Indexed: 12/22/2022] Open
Abstract
Cellular morphogenesis is governed by the prepattern based on the symmetry-breaking emergence of dense protein clusters. Thus, a cluster of active GTPase Cdc42 marks the site of nascent bud in the baker's yeast. An important biological question is which mechanisms control the number of pattern maxima (spots) and, thus, the number of nascent cellular structures. Distinct flavors of theoretical models seem to suggest different predictions. While the classical Turing scenario leads to an array of stably coexisting multiple structures, mass-conserved models predict formation of a single spot that emerges via the greedy competition between the pattern maxima for the common molecular resources. Both the outcome and the kinetics of this competition are of significant biological importance but remained poorly explored. Recent theoretical analyses largely addressed these questions, but their results have not yet been fully appreciated by the broad biological community. Keeping mathematical apparatus and jargon to the minimum, we review the main conclusions of these analyses with their biological implications in mind. Focusing on the specific example of pattern formation by small GTPases, we speculate on the features of the patterning mechanisms that bypass competition and favor formation of multiple coexisting structures and contrast them with those of the mechanisms that harness competition to form unique cellular structures.
Collapse
Affiliation(s)
- Andrew B. Goryachev
- SynthSys, Centre for Synthetic and Systems Biology, Institute for Cell Biology, University of Edinburgh, Edinburg EH9 3BD, UK;
| | | |
Collapse
|
211
|
Yoshino Y, Kobayashi A, Qi H, Endo S, Fang Z, Shindo K, Kanazawa R, Chiba N. RACK1 regulates centriole duplication through promoting the activation of polo-like kinase 1 by Aurora A. J Cell Sci 2020; 133:jcs238931. [PMID: 32788231 DOI: 10.1242/jcs.238931] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2019] [Accepted: 07/29/2020] [Indexed: 01/08/2023] Open
Abstract
Breast cancer gene 1 (BRCA1) contributes to the regulation of centrosome number. We previously identified receptor for activated C kinase 1 (RACK1) as a BRCA1-interacting partner. RACK1, a scaffold protein that interacts with multiple proteins through its seven WD40 domains, directly binds to BRCA1 and localizes to centrosomes. RACK1 knockdown suppresses centriole duplication, whereas RACK1 overexpression causes centriole overduplication in a subset of mammary gland-derived cells. In this study, we showed that RACK1 binds directly to polo-like kinase 1 (PLK1) and Aurora A, and promotes the Aurora A-PLK1 interaction. RACK1 knockdown decreased phosphorylated PLK1 (p-PLK1) levels and the centrosomal localization of Aurora A and p-PLK1 in S phase, whereas RACK1 overexpression increased p-PLK1 level and the centrosomal localization of Aurora A and p-PLK1 in interphase, resulting in an increase of cells with abnormal centriole disengagement. Overexpression of cancer-derived RACK1 variants failed to enhance the Aurora A-PLK1 interaction, PLK1 phosphorylation and the centrosomal localization of p-PLK1. These results suggest that RACK1 functions as a scaffold protein that promotes the activation of PLK1 by Aurora A in order to promote centriole duplication.This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Yuki Yoshino
- Department of Cancer Biology, Institute of Aging, Development, and Cancer, Tohoku University, 4-1 Seiryomachi, Aoba-ku, Sendai 980-8575, Japan
- Department of Cancer Biology, Tohoku University Graduate School of Medicine, 4-1 Seiryomachi Aoba-ku, Sendai 980-8575, Japan
- Laboratory of Cancer Biology, Graduate School of Life Sciences, Tohoku University, 4-1 Seiryomachi Aoba-ku, Sendai 980-8575, Japan
| | - Akihiro Kobayashi
- Department of Cancer Biology, Institute of Aging, Development, and Cancer, Tohoku University, 4-1 Seiryomachi, Aoba-ku, Sendai 980-8575, Japan
- Department of Cancer Biology, Tohoku University Graduate School of Medicine, 4-1 Seiryomachi Aoba-ku, Sendai 980-8575, Japan
| | - Huicheng Qi
- Department of Cancer Biology, Institute of Aging, Development, and Cancer, Tohoku University, 4-1 Seiryomachi, Aoba-ku, Sendai 980-8575, Japan
- Department of Cancer Biology, Tohoku University Graduate School of Medicine, 4-1 Seiryomachi Aoba-ku, Sendai 980-8575, Japan
| | - Shino Endo
- Department of Cancer Biology, Institute of Aging, Development, and Cancer, Tohoku University, 4-1 Seiryomachi, Aoba-ku, Sendai 980-8575, Japan
- Department of Cancer Biology, Tohoku University Graduate School of Medicine, 4-1 Seiryomachi Aoba-ku, Sendai 980-8575, Japan
| | - Zhenzhou Fang
- Department of Cancer Biology, Institute of Aging, Development, and Cancer, Tohoku University, 4-1 Seiryomachi, Aoba-ku, Sendai 980-8575, Japan
- Department of Cancer Biology, Tohoku University Graduate School of Medicine, 4-1 Seiryomachi Aoba-ku, Sendai 980-8575, Japan
| | - Kazuha Shindo
- Department of Cancer Biology, Institute of Aging, Development, and Cancer, Tohoku University, 4-1 Seiryomachi, Aoba-ku, Sendai 980-8575, Japan
- Laboratory of Cancer Biology, Graduate School of Life Sciences, Tohoku University, 4-1 Seiryomachi Aoba-ku, Sendai 980-8575, Japan
| | - Ryo Kanazawa
- Department of Cancer Biology, Institute of Aging, Development, and Cancer, Tohoku University, 4-1 Seiryomachi, Aoba-ku, Sendai 980-8575, Japan
- Laboratory of Cancer Biology, Graduate School of Life Sciences, Tohoku University, 4-1 Seiryomachi Aoba-ku, Sendai 980-8575, Japan
| | - Natsuko Chiba
- Department of Cancer Biology, Institute of Aging, Development, and Cancer, Tohoku University, 4-1 Seiryomachi, Aoba-ku, Sendai 980-8575, Japan
- Department of Cancer Biology, Tohoku University Graduate School of Medicine, 4-1 Seiryomachi Aoba-ku, Sendai 980-8575, Japan
- Laboratory of Cancer Biology, Graduate School of Life Sciences, Tohoku University, 4-1 Seiryomachi Aoba-ku, Sendai 980-8575, Japan
| |
Collapse
|
212
|
Ong JY, Torres JZ. Phase Separation in Cell Division. Mol Cell 2020; 80:9-20. [PMID: 32860741 DOI: 10.1016/j.molcel.2020.08.007] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Revised: 07/10/2020] [Accepted: 08/07/2020] [Indexed: 02/08/2023]
Abstract
Cell division requires the assembly and organization of a microtubule spindle for the proper separation of chromosomes in mitosis and meiosis. Phase separation is an emerging paradigm for understanding spatial and temporal regulation of a variety of cellular processes, including cell division. Phase-separated condensates have been recently discovered at many structures during cell division as a possible mechanism for properly localizing, organizing, and activating proteins involved in cell division. Here, we review how these condensates play roles in regulating microtubule density and organization and spindle assembly and function and in activating some of the key players in cell division. We conclude with perspectives on areas of future research for this exciting and rapidly advancing field.
Collapse
Affiliation(s)
- Joseph Y Ong
- Department of Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Jorge Z Torres
- Department of Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, CA 90095, USA; Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA; Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| |
Collapse
|
213
|
Brigant B, Demont Y, Ouled-Haddou H, Metzinger-Le Meuth V, Testelin S, Garçon L, Metzinger L, Rochette J. TRIM37 is highly expressed during mitosis in CHON-002 chondrocytes cell line and is regulated by miR-223. Bone 2020; 137:115393. [PMID: 32353567 DOI: 10.1016/j.bone.2020.115393] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 04/24/2020] [Accepted: 04/24/2020] [Indexed: 02/06/2023]
Abstract
Multiple molecular disorders can affect mechanisms regulating proliferation and differentiation of growth plate chondrocytes. Mutations in the TRIM37 gene cause the Mulibrey nanism, a heritable growth disorder. Since chondrocytes are instrumental in long bone growth that is deficient in nanism, we hypothesized that TRIM37 defect could contribute to dysregulation of the chondrocyte cell cycle. Western blotting, confocal microscopy and imaging flow cytometry determined TRIM37 expression in CHON-002 cell lineage. We showed that TRIM37 is expressed during mitosis of chondrocytes and directly impacted their proliferation. During the chondrocyte cell cycle, TRIM37 was present in both nucleus and cytoplasm. During M phase we observed an increase of the TRIM37-Tubulin co-localization in comparison with G1, S and G2 phases. TRIM37 knock down inhibited proliferation, together with cell cycle anomalies and increased autophagy, while overexpression accordingly enhanced cell proliferation. We demonstrated that microRNA-223 directly targets TRIM37, and suggest that miR-223 regulates TRIM37 gene expression during the cell cycle. In summary, our results give clues to explain why TRIM37 deficiency in chondrocytes impacts bone growth. Modulating TRIM37 using miR-223 could be an approach to increase chondrogenesis.
Collapse
Affiliation(s)
- Benjamin Brigant
- HEMATIM EA4666, Centre Universitaire de Recherche en Santé, Université de Picardie Jules Verne, Amiens, France
| | - Yohann Demont
- HEMATIM EA4666, Centre Universitaire de Recherche en Santé, Université de Picardie Jules Verne, Amiens, France
| | - Hakim Ouled-Haddou
- HEMATIM EA4666, Centre Universitaire de Recherche en Santé, Université de Picardie Jules Verne, Amiens, France
| | | | - Sylvie Testelin
- Maxillo-Facial Surgery Department, Centre Hospitalo-Universitaire d'Amiens, Avenue Laennec, 80000 Amiens, France; EA CHIMERE, université de Picardie-Jules-Verne, Avenue Laennec, 80000 Amiens, France; Facing Faces Institute, Avenue Laennec, 80000 Amiens, France
| | - Loïc Garçon
- HEMATIM EA4666, Centre Universitaire de Recherche en Santé, Université de Picardie Jules Verne, Amiens, France
| | - Laurent Metzinger
- HEMATIM EA4666, Centre Universitaire de Recherche en Santé, Université de Picardie Jules Verne, Amiens, France
| | - Jacques Rochette
- HEMATIM EA4666, Centre Universitaire de Recherche en Santé, Université de Picardie Jules Verne, Amiens, France.
| |
Collapse
|
214
|
Lee KS, Park JE, Il Ahn J, Wei Z, Zhang L. A self-assembled cylindrical platform for Plk4-induced centriole biogenesis. Open Biol 2020; 10:200102. [PMID: 32810424 PMCID: PMC7479937 DOI: 10.1098/rsob.200102] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Accepted: 05/28/2020] [Indexed: 12/19/2022] Open
Abstract
The centrosome, a unique membraneless multiprotein organelle, plays a pivotal role in various cellular processes that are critical for promoting cell proliferation. Faulty assembly or organization of the centrosome results in abnormal cell division, which leads to various human disorders including cancer, microcephaly and ciliopathy. Recent studies have provided new insights into the stepwise self-assembly of two pericentriolar scaffold proteins, Cep63 and Cep152, into a near-micrometre-scale higher-order structure whose architectural properties could be crucial for proper execution of its biological function. The construction of the scaffold architecture appears to be centrally required for tight control of a Ser/Thr kinase called Plk4, a key regulator of centriole duplication, which occurs precisely once per cell cycle. In this review, we will discuss a new paradigm for understanding how pericentrosomal scaffolds are self-organized into a new functional entity and how, on the resulting structural platform, Plk4 undergoes physico-chemical conversion to trigger centriole biogenesis.
Collapse
Affiliation(s)
- Kyung S. Lee
- Laboratory of Metabolism, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | | | | |
Collapse
|
215
|
Luan W, Hao CZ, Li JQ, Wei Q, Gong JY, Qiu YL, Lu Y, Shen CH, Xia Q, Xie XB, Zhang MH, Abuduxikuer K, Li ZD, Wang L, Xing QH, Knisely AS, Wang JS. Biallelic loss-of-function ZFYVE19 mutations are associated with congenital hepatic fibrosis, sclerosing cholangiopathy and high-GGT cholestasis. J Med Genet 2020; 58:514-525. [PMID: 32737136 DOI: 10.1136/jmedgenet-2019-106706] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2019] [Revised: 05/28/2020] [Accepted: 06/20/2020] [Indexed: 12/14/2022]
Abstract
BACKGROUND For many children with intrahepatic cholestasis and high-serum gamma-glutamyl transferase (GGT) activity, a genetic aetiology of hepatobiliary disease remains undefined. We sought to identify novel genes mutated in children with idiopathic high-GGT intrahepatic cholestasis, with clinical, histopathological and functional correlations. METHODS We assembled a cohort of 25 children with undiagnosed high-GGT cholestasis and without clinical features of biliary-tract infection or radiological features of choledochal malformation, sclerosing cholangitis or cholelithiasis. Mutations were identified through whole-exome sequencing and targeted Sanger sequencing. We reviewed histopathological findings and assessed phenotypical effects of ZFYVE19 deficiency in cultured cells by immunofluorescence microscopy. RESULTS Nine Han Chinese children harboured biallelic, predictedly complete loss-of-function pathogenic mutations in ZFYVE19 (c.314C>G, p.S105X; c.379C>T, p.Q127X; c.514C>T, p.R172X; c.547C>T, p.R183X; c.226A>G, p.M76V). All had portal hypertension and, at liver biopsy, histopathological features of the ductal plate malformation (DPM)/congenital hepatic fibrosis (CHF). Four children required liver transplantation for recurrent gastrointestinal haemorrhage. DPM/CHF was confirmed at hepatectomy, with sclerosing small-duct cholangitis. Immunostaining for two primary-cilium axonemal proteins found expression that was deficient intraluminally and ectopic within cholangiocyte cytoplasm. ZFYVE19 depletion in cultured cells yielded abnormalities of centriole and axoneme. CONCLUSION Biallelic ZFYVE19 mutations can lead to high-GGT cholestasis and DPM/CHF in vivo. In vitro, they can lead to centriolar and axonemal abnormalities. These observations indicate that mutation in ZFYVE19 results, through as yet undefined mechanisms, in a ciliopathy.
Collapse
Affiliation(s)
- Weisha Luan
- Department of Pediatrics, Jinshan Hospital of Fudan University, Shanghai, China.,The Center for Pediatric Liver Diseases, Children's Hospital of Fudan University, Shanghai, China
| | - Chen-Zhi Hao
- The Center for Pediatric Liver Diseases, Children's Hospital of Fudan University, Shanghai, China
| | - Jia-Qi Li
- Department of Pediatrics, Jinshan Hospital of Fudan University, Shanghai, China.,The Center for Pediatric Liver Diseases, Children's Hospital of Fudan University, Shanghai, China
| | - Qing Wei
- Laboratory for Reproductive Health, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, China
| | - Jing-Yu Gong
- Department of Pediatrics, Jinshan Hospital of Fudan University, Shanghai, China
| | - Yi-Ling Qiu
- Department of Pediatrics, Jinshan Hospital of Fudan University, Shanghai, China.,The Center for Pediatric Liver Diseases, Children's Hospital of Fudan University, Shanghai, China
| | - Yi Lu
- The Center for Pediatric Liver Diseases, Children's Hospital of Fudan University, Shanghai, China
| | - Cong-Huan Shen
- Department of Surgery, Huashan Hospital Affiliated to Fudan University, Shanghai, China
| | - Qiang Xia
- Department of Liver Surgery and Liver Transplantation Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xin-Bao Xie
- The Center for Pediatric Liver Diseases, Children's Hospital of Fudan University, Shanghai, China
| | - Mei-Hong Zhang
- Department of Pediatrics, Jinshan Hospital of Fudan University, Shanghai, China
| | - Kuerbanjiang Abuduxikuer
- The Center for Pediatric Liver Diseases, Children's Hospital of Fudan University, Shanghai, China
| | - Zhong-Die Li
- The Center for Pediatric Liver Diseases, Children's Hospital of Fudan University, Shanghai, China
| | - Li Wang
- The Center for Pediatric Liver Diseases, Children's Hospital of Fudan University, Shanghai, China
| | - Qing-He Xing
- Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - A S Knisely
- Institut für Pathologie, Medizinische Universität Graz, Graz, Austria
| | - Jian-She Wang
- The Center for Pediatric Liver Diseases, Children's Hospital of Fudan University, Shanghai, China
| |
Collapse
|
216
|
Park EM, Scott PM, Clutario K, Cassidy KB, Zhan K, Gerber SA, Holland AJ. WBP11 is required for splicing the TUBGCP6 pre-mRNA to promote centriole duplication. J Cell Biol 2020; 219:133543. [PMID: 31874114 PMCID: PMC7039186 DOI: 10.1083/jcb.201904203] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 09/24/2019] [Accepted: 10/22/2019] [Indexed: 12/15/2022] Open
Abstract
Centriole duplication occurs once in each cell cycle to maintain centrosome number. A previous genome-wide screen revealed that depletion of 14 RNA splicing factors leads to a specific defect in centriole duplication, but the cause of this deficit remains unknown. Here, we identified an additional pre-mRNA splicing factor, WBP11, as a novel protein required for centriole duplication. Loss of WBP11 results in the retention of ∼200 introns, including multiple introns in TUBGCP6, a central component of the γ-TuRC. WBP11 depletion causes centriole duplication defects, in part by causing a rapid decline in the level of TUBGCP6. Several additional splicing factors that are required for centriole duplication interact with WBP11 and are required for TUBGCP6 expression. These findings provide insight into how the loss of a subset of splicing factors leads to a failure of centriole duplication. This may have clinical implications because mutations in some spliceosome proteins cause microcephaly and/or growth retardation, phenotypes that are strongly linked to centriole defects.
Collapse
Affiliation(s)
- Elizabeth M Park
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Phillip M Scott
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Kevin Clutario
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Katelyn B Cassidy
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Lebanon, NH
| | - Kevin Zhan
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Scott A Gerber
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Lebanon, NH.,Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Lebanon, NH.,Norris Cotton Cancer Center, Dartmouth-Hitchcock Medical Center, Lebanon, NH
| | - Andrew J Holland
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD
| |
Collapse
|
217
|
Otsuka K, Yoshino Y, Qi H, Chiba N. The Function of BARD1 in Centrosome Regulation in Cooperation with BRCA1/OLA1/RACK1. Genes (Basel) 2020; 11:genes11080842. [PMID: 32722046 PMCID: PMC7464954 DOI: 10.3390/genes11080842] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 07/21/2020] [Accepted: 07/22/2020] [Indexed: 12/17/2022] Open
Abstract
Breast cancer gene 1 (BRCA1)-associated RING domain protein 1 (BARD1) forms a heterodimer with BRCA1, a tumor suppressor associated with hereditary breast and ovarian cancer. BRCA1/BARD1 functions in multiple cellular processes including DNA repair and centrosome regulation. Centrosomes are the major microtubule-organizing centers in animal cells and are critical for the formation of a bipolar mitotic spindle. BRCA1 and BARD1 localize to the centrosome during the cell cycle, and the BRCA1/BARD1 dimer ubiquitinates centrosomal proteins to regulate centrosome function. We identified Obg-like ATPase 1 (OLA1) and receptor for activated C kinase (RACK1) as BRCA1/BARD1-interating proteins that bind to BARD1 and BRCA1 and localize the centrosomes during the cell cycle. Cancer-derived variants of BRCA1, BARD1, OLA1, and RACK1 failed to interact, and aberrant expression of these proteins caused centrosome amplification due to centriole overduplication only in mammary tissue-derived cells. In S-G2 phase, the number of centrioles was higher in mammary tissue-derived cells than in cells from other tissues, suggesting their involvement in tissue-specific carcinogenesis by BRCA1 and BARD1 germline mutations. We described the function of BARD1 in centrosome regulation in cooperation with BRCA1/OLA1/RACK1, as well as the effect of their dysfunction on carcinogenesis.
Collapse
Affiliation(s)
- Kei Otsuka
- Department of Cancer Biology, Institute of Development, Aging and Cancer (IDAC), Tohoku University, 4-1 Seiryomachi Aoba-ku, Sendai 980-8575, Japan; (K.O.); (Y.Y.); (H.Q.)
- Laboratory of Cancer Biology, Graduate School of Life Sciences, Tohoku University, 4-1 Seiryomachi Aoba-ku, Sendai 980-8575, Japan
| | - Yuki Yoshino
- Department of Cancer Biology, Institute of Development, Aging and Cancer (IDAC), Tohoku University, 4-1 Seiryomachi Aoba-ku, Sendai 980-8575, Japan; (K.O.); (Y.Y.); (H.Q.)
- Laboratory of Cancer Biology, Graduate School of Life Sciences, Tohoku University, 4-1 Seiryomachi Aoba-ku, Sendai 980-8575, Japan
- Department of Cancer Biology, Tohoku University Graduate School of Medicine, 4-1 Seiryomachi Aoba-ku, Sendai 980-8575, Japan
| | - Huicheng Qi
- Department of Cancer Biology, Institute of Development, Aging and Cancer (IDAC), Tohoku University, 4-1 Seiryomachi Aoba-ku, Sendai 980-8575, Japan; (K.O.); (Y.Y.); (H.Q.)
- Department of Cancer Biology, Tohoku University Graduate School of Medicine, 4-1 Seiryomachi Aoba-ku, Sendai 980-8575, Japan
| | - Natsuko Chiba
- Department of Cancer Biology, Institute of Development, Aging and Cancer (IDAC), Tohoku University, 4-1 Seiryomachi Aoba-ku, Sendai 980-8575, Japan; (K.O.); (Y.Y.); (H.Q.)
- Laboratory of Cancer Biology, Graduate School of Life Sciences, Tohoku University, 4-1 Seiryomachi Aoba-ku, Sendai 980-8575, Japan
- Department of Cancer Biology, Tohoku University Graduate School of Medicine, 4-1 Seiryomachi Aoba-ku, Sendai 980-8575, Japan
- Correspondence:
| |
Collapse
|
218
|
Liu X, Liu X, Wang H, Dou Z, Ruan K, Hill DL, Li L, Shi Y, Yao X. Phase separation drives decision making in cell division. J Biol Chem 2020; 295:13419-13431. [PMID: 32699013 DOI: 10.1074/jbc.rev120.011746] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 07/22/2020] [Indexed: 12/11/2022] Open
Abstract
Liquid-liquid phase separation (LLPS) of biomolecules drives the formation of subcellular compartments with distinct physicochemical properties. These compartments, free of lipid bilayers and therefore called membraneless organelles, include nucleoli, centrosomes, heterochromatin, and centromeres. These have emerged as a new paradigm to account for subcellular organization and cell fate decisions. Here we summarize recent studies linking LLPS to mitotic spindle, heterochromatin, and centromere assembly and their plasticity controls in the context of the cell division cycle, highlighting a functional role for phase behavior and material properties of proteins assembled onto heterochromatin, centromeres, and central spindles via LLPS. The techniques and tools for visualizing and harnessing membraneless organelle dynamics and plasticity in mitosis are also discussed, as is the potential for these discoveries to promote new research directions for investigating chromosome dynamics, plasticity, and interchromosome interactions in the decision-making process during mitosis.
Collapse
Affiliation(s)
- Xing Liu
- MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics and CAS Center for Excellence in Molecular Cell Science, University of Science and Technology of China School of Life Science, Hefei, China; Anhui Key Laboratory for Cellular Dynamics & Chemical Biology, Hefei National Center for Physical Sciences at Nanoscale, Hefei, China; Keck Center for Cellular Dynamics and Organoids Plasticity, Morehouse School of Medicine, Atlanta, Georgia, USA
| | - Xu Liu
- MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics and CAS Center for Excellence in Molecular Cell Science, University of Science and Technology of China School of Life Science, Hefei, China; Anhui Key Laboratory for Cellular Dynamics & Chemical Biology, Hefei National Center for Physical Sciences at Nanoscale, Hefei, China; Keck Center for Cellular Dynamics and Organoids Plasticity, Morehouse School of Medicine, Atlanta, Georgia, USA
| | - Haowei Wang
- MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics and CAS Center for Excellence in Molecular Cell Science, University of Science and Technology of China School of Life Science, Hefei, China; Anhui Key Laboratory for Cellular Dynamics & Chemical Biology, Hefei National Center for Physical Sciences at Nanoscale, Hefei, China
| | - Zhen Dou
- MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics and CAS Center for Excellence in Molecular Cell Science, University of Science and Technology of China School of Life Science, Hefei, China; Anhui Key Laboratory for Cellular Dynamics & Chemical Biology, Hefei National Center for Physical Sciences at Nanoscale, Hefei, China
| | - Ke Ruan
- MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics and CAS Center for Excellence in Molecular Cell Science, University of Science and Technology of China School of Life Science, Hefei, China; Anhui Key Laboratory for Cellular Dynamics & Chemical Biology, Hefei National Center for Physical Sciences at Nanoscale, Hefei, China
| | - Donald L Hill
- Comprehensive Cancer Center, University of Alabama, Birmingham, Alabama, USA
| | - Lin Li
- CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Shanghai, China
| | - Yunyu Shi
- MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics and CAS Center for Excellence in Molecular Cell Science, University of Science and Technology of China School of Life Science, Hefei, China; Anhui Key Laboratory for Cellular Dynamics & Chemical Biology, Hefei National Center for Physical Sciences at Nanoscale, Hefei, China
| | - Xuebiao Yao
- MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics and CAS Center for Excellence in Molecular Cell Science, University of Science and Technology of China School of Life Science, Hefei, China; Anhui Key Laboratory for Cellular Dynamics & Chemical Biology, Hefei National Center for Physical Sciences at Nanoscale, Hefei, China; Keck Center for Cellular Dynamics and Organoids Plasticity, Morehouse School of Medicine, Atlanta, Georgia, USA; Comprehensive Cancer Center, University of Alabama, Birmingham, Alabama, USA; CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Shanghai, China.
| |
Collapse
|
219
|
Abstract
Drosophila spermatocyte centrioles are ideal for imaging studies. Their large, characteristic V conformation is both easy to identify and measure using standard imaging techniques. However, certain detailed features, such as their ninefold symmetry, are only visible below the diffraction limit of light. This is therefore a system that can benefit from the increased effective resolution potentially achievable by expansion microscopy. Here, I provide detailed protocols of two types of expansion microscopy methodologies applied to Drosophila spermatocyte centrioles, and discuss which is able to achieve the highest effective resolution in this system. I describe how to precisely measure these organelles post-expansion, and discuss how they can therefore be used as "molecular rulers" to troubleshoot and compare expansion techniques. I also provide protocols to combine expansion microscopy with super-resolution imaging in this tissue, discussing potential pitfalls. I conclude that expansion microscopy provides an effective alternative for thick tissues that are not amenable for traditional super-resolution techniques.
Collapse
Affiliation(s)
- Alan Wainman
- Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom.
| |
Collapse
|
220
|
Zhao H, Yang S, Chen Q, Duan X, Li G, Huang Q, Zhu X, Yan X. Cep57 and Cep57l1 function redundantly to recruit the Cep63-Cep152 complex for centriole biogenesis. J Cell Sci 2020; 133:jcs241836. [PMID: 32503940 DOI: 10.1242/jcs.241836] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Accepted: 05/27/2020] [Indexed: 12/30/2022] Open
Abstract
The Cep63-Cep152 complex located at the mother centriole recruits Plk4 to initiate centriole biogenesis. How the complex is targeted to mother centrioles, however, is unclear. In this study, we show that Cep57 and its paralog, Cep57l1, colocalize with Cep63 and Cep152 at the proximal end of mother centrioles in both cycling cells and multiciliated cells undergoing centriole amplification. Both Cep57 and Cep57l1 bind to the centrosomal targeting region of Cep63. The depletion of both proteins, but not either one, blocks loading of the Cep63-Cep152 complex to mother centrioles and consequently prevents centriole duplication. We propose that Cep57 and Cep57l1 function redundantly to ensure recruitment of the Cep63-Cep152 complex to the mother centrioles for procentriole formation.
Collapse
Affiliation(s)
- Huijie Zhao
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China
| | - Sen Yang
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Qingxia Chen
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- School of Life Science and Technology, ShanghaiTech University, 100 Haike Road, Shanghai 201210, China
| | - Xiaomeng Duan
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China
| | - Guoqing Li
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China
| | - Qiongping Huang
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China
| | - Xueliang Zhu
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- School of Life Science and Technology, ShanghaiTech University, 100 Haike Road, Shanghai 201210, China
| | - Xiumin Yan
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China
| |
Collapse
|
221
|
Osman MA, Antonisamy WJ, Yakirevich E. IQGAP1 control of centrosome function defines distinct variants of triple negative breast cancer. Oncotarget 2020; 11:2493-2511. [PMID: 32655836 PMCID: PMC7335670 DOI: 10.18632/oncotarget.27623] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Accepted: 05/14/2020] [Indexed: 12/12/2022] Open
Abstract
Triple negative breast cancer (TNBC) is a heterogenous and lethal disease that lacks diagnostic markers and therapeutic targets; as such common targets are highly sought after. IQGAP1 is a signaling scaffold implicated in TNBC, but its mechanism is unknown. Here we show that IQGAP1 localizes to the centrosome, interacts with and influences the expression level and localization of key centrosome proteins like BRCA1 and thereby impacts centrosome number. Genetic mutant analyses suggest that phosphorylation cycling of IQGAP1 is important to its subcellular localization and centrosome-nuclear shuttling of BRCA1; dysfunction of this process defines two alternate mechanisms associated with cell proliferation. TNBC cell lines and patient tumor tissues differentially phenocopy these mechanisms supporting clinical existence of molecularly distinct variants of TNBC defined by IQGAP1 pathways. These variants are defined, at least in part, by differential mis-localization or stabilization of IQGAP1-BRCA1 and rewiring of a novel Erk1/2-MNK1-JNK-Akt-β-catenin signaling signature. We discuss a model in which IQGAP1 modulates centrosome-nuclear crosstalk to regulate cell division and imparts on cancer. These findings have implications on cancer racial disparities and can provide molecular tools for classification of TNBC, presenting IQGAP1 as a common target amenable to personalized medicine.
Collapse
Affiliation(s)
- Mahasin A. Osman
- Department of Medicine, Division of Oncology, Health Sciences Campus, University of Toledo, Toledo, OH 43614, USA
- Department of Molecular Pharmacology, Physiology and Biotechnology, Division of Biology and Medicine, Warren Alpert Medical School of Brown University, Providence, RI 02912, USA
| | - William James Antonisamy
- Department of Medicine, Division of Oncology, Health Sciences Campus, University of Toledo, Toledo, OH 43614, USA
| | - Evgeny Yakirevich
- Department of Pathology and Laboratory Medicine, Rhode Island Hospital, Warren Alpert Medical School of Brown University, Providence, RI 02903, USA
| |
Collapse
|
222
|
Lin YN, Lee YS, Li SK, Tang TK. Loss of CPAP in developing mouse brain and its functional implication for human primary microcephaly. J Cell Sci 2020; 133:jcs243592. [PMID: 32501282 DOI: 10.1242/jcs.243592] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Accepted: 05/07/2020] [Indexed: 12/21/2022] Open
Abstract
Primary microcephaly (MCPH) is a neurodevelopmental disorder characterized by small brain size with mental retardation. CPAP (also known as CENPJ), a known microcephaly-associated gene, plays a key role in centriole biogenesis. Here, we generated a previously unreported conditional knockout allele in the mouse Cpap gene. Our results showed that conditional Cpap deletion in the central nervous system preferentially induces formation of monopolar spindles in radial glia progenitors (RGPs) at around embryonic day 14.5 and causes robust apoptosis that severely disrupts embryonic brains. Interestingly, microcephalic brains with reduced apoptosis are detected in conditional Cpap gene-deleted mice that lose only one allele of p53 (also known as Trp53), while simultaneous removal of p53 and Cpap rescues RGP death. Furthermore, Cpap deletion leads to cilia loss, RGP mislocalization, junctional integrity disruption, massive heterotopia and severe cerebellar hypoplasia. Together, these findings indicate that complete CPAP loss leads to severe and complex phenotypes in developing mouse brain, and provide new insights into the causes of MCPH.
Collapse
Affiliation(s)
- Yi-Nan Lin
- Institute of Biomedical Sciences, Academia Sinica, Taipei, 11529 Taiwan
| | - Ying-Shan Lee
- Institute of Biomedical Sciences, Academia Sinica, Taipei, 11529 Taiwan
| | - Shu-Kuei Li
- Institute of Biomedical Sciences, Academia Sinica, Taipei, 11529 Taiwan
| | - Tang K Tang
- Institute of Biomedical Sciences, Academia Sinica, Taipei, 11529 Taiwan
| |
Collapse
|
223
|
Gartenmann L, Vicente CC, Wainman A, Novak ZA, Sieber B, Richens JH, Raff JW. Drosophila Sas-6, Ana2 and Sas-4 self-organise into macromolecular structures that can be used to probe centriole and centrosome assembly. J Cell Sci 2020; 133:jcs244574. [PMID: 32409564 PMCID: PMC7328145 DOI: 10.1242/jcs.244574] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Accepted: 04/24/2020] [Indexed: 01/02/2023] Open
Abstract
Centriole assembly requires a small number of conserved proteins. The precise pathway of centriole assembly has been difficult to study, as the lack of any one of the core assembly proteins [Plk4, Ana2 (the homologue of mammalian STIL), Sas-6, Sas-4 (mammalian CPAP) or Asl (mammalian Cep152)] leads to the absence of centrioles. Here, we use Sas-6 and Ana2 particles (SAPs) as a new model to probe the pathway of centriole and centrosome assembly. SAPs form in Drosophila eggs or embryos when Sas-6 and Ana2 are overexpressed. SAP assembly requires Sas-4, but not Plk4, whereas Asl helps to initiate SAP assembly but is not required for SAP growth. Although not centrioles, SAPs recruit and organise many centriole and centrosome components, nucleate microtubules, organise actin structures and compete with endogenous centrosomes to form mitotic spindle poles. SAPs require Asl to efficiently recruit pericentriolar material (PCM), but Spd-2 (the homologue of mammalian Cep192) can promote some PCM assembly independently of Asl. These observations provide new insights into the pathways of centriole and centrosome assembly.
Collapse
Affiliation(s)
- Lisa Gartenmann
- Sir William Dunn School of Pathology, University of Oxford, South Parks Rd, Oxford OX1 3RE, UK
| | - Catarina C Vicente
- Sir William Dunn School of Pathology, University of Oxford, South Parks Rd, Oxford OX1 3RE, UK
| | - Alan Wainman
- Sir William Dunn School of Pathology, University of Oxford, South Parks Rd, Oxford OX1 3RE, UK
| | - Zsofi A Novak
- Sir William Dunn School of Pathology, University of Oxford, South Parks Rd, Oxford OX1 3RE, UK
| | - Boris Sieber
- Sir William Dunn School of Pathology, University of Oxford, South Parks Rd, Oxford OX1 3RE, UK
| | - Jennifer H Richens
- Sir William Dunn School of Pathology, University of Oxford, South Parks Rd, Oxford OX1 3RE, UK
| | - Jordan W Raff
- Sir William Dunn School of Pathology, University of Oxford, South Parks Rd, Oxford OX1 3RE, UK
| |
Collapse
|
224
|
Bozal-Basterra L, Gonzalez-Santamarta M, Muratore V, Bermejo-Arteagabeitia A, Da Fonseca C, Barroso-Gomila O, Azkargorta M, Iloro I, Pampliega O, Andrade R, Martín-Martín N, Branon TC, Ting AY, Rodríguez JA, Carracedo A, Elortza F, Sutherland JD, Barrio R. LUZP1, a novel regulator of primary cilia and the actin cytoskeleton, is a contributing factor in Townes-Brocks Syndrome. eLife 2020; 9:e55957. [PMID: 32553112 PMCID: PMC7363444 DOI: 10.7554/elife.55957] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Accepted: 06/18/2020] [Indexed: 12/20/2022] Open
Abstract
Primary cilia are sensory organelles crucial for cell signaling during development and organ homeostasis. Cilia arise from centrosomes and their formation and function is governed by numerous factors. Through our studies on Townes-Brocks Syndrome (TBS), a rare disease linked to abnormal cilia formation in human fibroblasts, we uncovered the leucine-zipper protein LUZP1 as an interactor of truncated SALL1, a dominantly-acting protein causing the disease. Using TurboID proximity labeling and pulldowns, we show that LUZP1 associates with factors linked to centrosome and actin filaments. Here, we show that LUZP1 is a cilia regulator. It localizes around the centrioles and to actin cytoskeleton. Loss of LUZP1 reduces F-actin levels, facilitates ciliogenesis and alters Sonic Hedgehog signaling, pointing to a key role in cytoskeleton-cilia interdependency. Truncated SALL1 increases the ubiquitin proteasome-mediated degradation of LUZP1. Together with other factors, alterations in LUZP1 may be contributing to TBS etiology.
Collapse
Affiliation(s)
- Laura Bozal-Basterra
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology ParkDerioSpain
| | - María Gonzalez-Santamarta
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology ParkDerioSpain
| | - Veronica Muratore
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology ParkDerioSpain
| | - Aitor Bermejo-Arteagabeitia
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology ParkDerioSpain
| | - Carolina Da Fonseca
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology ParkDerioSpain
| | - Orhi Barroso-Gomila
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology ParkDerioSpain
| | - Mikel Azkargorta
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology ParkDerioSpain
- CIBERehd, Instituto de Salud Carlos IIIMadridSpain
- ProteoRed-ISCIII, Instituto de Salud Carlos IIIMadridSpain
| | - Ibon Iloro
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology ParkDerioSpain
- CIBERehd, Instituto de Salud Carlos IIIMadridSpain
- ProteoRed-ISCIII, Instituto de Salud Carlos IIIMadridSpain
| | - Olatz Pampliega
- Department of Neurosciences, University of the Basque Country, Achucarro Basque Center for Neuroscience-UPV/EHULeioaSpain
| | - Ricardo Andrade
- Analytical & High Resolution Biomedical Microscopy Core Facility, University of the Basque Country (UPV/EHU)LeioaSpain
| | - Natalia Martín-Martín
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology ParkDerioSpain
| | - Tess C Branon
- Department of Chemistry, Massachusetts Institute of TechnologyCambridgeUnited States
- Departments of Genetics, Chemistry and Biology, Stanford UniversityStanfordUnited States
| | - Alice Y Ting
- Department of Chemistry, Massachusetts Institute of TechnologyCambridgeUnited States
- Departments of Genetics, Chemistry and Biology, Stanford UniversityStanfordUnited States
- Chan Zuckerberg BiohubSan FranciscoUnited States
| | - Jose A Rodríguez
- Department of Genetics, Physical Anthropology and Animal Physiology, University of the Basque Country (UPV/EHU)LeioaSpain
| | - Arkaitz Carracedo
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology ParkDerioSpain
- CIBERONC, Instituto de Salud Carlos IIIMadridSpain
- Ikerbasque, Basque Foundation for ScienceBilbaoSpain
- Biochemistry and Molecular Biology Department, University of the Basque Country (UPV/EHU)BilbaoSpain
| | - Felix Elortza
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology ParkDerioSpain
- CIBERehd, Instituto de Salud Carlos IIIMadridSpain
- ProteoRed-ISCIII, Instituto de Salud Carlos IIIMadridSpain
| | - James D Sutherland
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology ParkDerioSpain
| | - Rosa Barrio
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology ParkDerioSpain
| |
Collapse
|
225
|
Functional genetic variants in centrosome-related genes CEP72 and YWHAG confer susceptibility to gastric cancer. Arch Toxicol 2020; 94:2861-2872. [PMID: 32535685 DOI: 10.1007/s00204-020-02782-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2019] [Accepted: 05/07/2020] [Indexed: 01/06/2023]
Abstract
Structural and numeric centrosome aberrations can induce chromosome segregation errors and promote tumor development and progression. We systematically evaluated associations of 19,603 single nucleotide polymorphisms (SNPs) across 136 centrosome-related genes with gastric cancer (GC) risk using four GWAS datasets with a total of 3771 cases and 5426 controls. We identified two loci at 15p13.3 and 7q11.23 significantly associated with GC risk, whose risk alleles were correlated with increased mRNA expression of CEP72 (P = 7.30 × 10-4) and YWHAG (P = 1.60 × 10-3), respectively. Dual-luciferase reporter assays confirmed that the risk T allele of rs924607 at 15p13.3 significantly increased a promoter activity of the reporter gene, leading to a higher CEP72 expression level. At 7q11.23, the risk haplotype of rs2961037 [G]-rs2961038 [G] significantly elevated an enhancer activity and the expression of YWHAG. Both the mRNA and protein levels of CEP72 and YWHAG were overexpressed in GC tumor tissues compared with peritumor tissues and overexpression of either gene showed an unfavorable prognosis of GC patients. Moreover, knockdown of either CEP72 or YWHAG inhibited GC cell proliferation, migration and invasion and promoted GC cell apoptosis. The genes coexpressed with CEP72 or YWHAG in GC tumor tissues were enriched in the Ras signaling pathway, which was confirmed that knockdown of either one decreased the expression of cyclin D1 but increased the expression of p21 and p27. In conclusion, genetic variants at 15p13.3 and 7q11.23 may confer GC risk via modulating the biological functions of CEP72 and YWHAG, respectively, suggesting the importance of centrosome-regulated genes in GC development.
Collapse
|
226
|
Aydogan MG, Steinacker TL, Mofatteh M, Wilmott ZM, Zhou FY, Gartenmann L, Wainman A, Saurya S, Novak ZA, Wong SS, Goriely A, Boemo MA, Raff JW. An Autonomous Oscillation Times and Executes Centriole Biogenesis. Cell 2020; 181:1566-1581.e27. [PMID: 32531200 PMCID: PMC7327525 DOI: 10.1016/j.cell.2020.05.018] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Revised: 12/19/2019] [Accepted: 05/08/2020] [Indexed: 01/18/2023]
Abstract
The accurate timing and execution of organelle biogenesis is crucial for cell physiology. Centriole biogenesis is regulated by Polo-like kinase 4 (Plk4) and initiates in S-phase when a daughter centriole grows from the side of a pre-existing mother. Here, we show that a Plk4 oscillation at the base of the growing centriole initiates and times centriole biogenesis to ensure that centrioles grow at the right time and to the right size. The Plk4 oscillation is normally entrained to the cell-cycle oscillator but can run autonomously of it-potentially explaining why centrioles can duplicate independently of cell-cycle progression. Mathematical modeling indicates that the Plk4 oscillation can be generated by a time-delayed negative feedback loop in which Plk4 inactivates the interaction with its centriolar receptor through multiple rounds of phosphorylation. We hypothesize that similar organelle-specific oscillations could regulate the timing and execution of organelle biogenesis more generally.
Collapse
Affiliation(s)
- Mustafa G Aydogan
- Sir William Dunn School of Pathology, University of Oxford, Oxford OX1 3RE, UK.
| | - Thomas L Steinacker
- Sir William Dunn School of Pathology, University of Oxford, Oxford OX1 3RE, UK
| | - Mohammad Mofatteh
- Sir William Dunn School of Pathology, University of Oxford, Oxford OX1 3RE, UK
| | - Zachary M Wilmott
- Sir William Dunn School of Pathology, University of Oxford, Oxford OX1 3RE, UK; Mathematical Institute, University of Oxford, Oxford OX2 6GG, UK
| | - Felix Y Zhou
- Ludwig Institute for Cancer Research, University of Oxford, Oxford OX3 7DQ, UK
| | - Lisa Gartenmann
- Sir William Dunn School of Pathology, University of Oxford, Oxford OX1 3RE, UK
| | - Alan Wainman
- Sir William Dunn School of Pathology, University of Oxford, Oxford OX1 3RE, UK
| | - Saroj Saurya
- Sir William Dunn School of Pathology, University of Oxford, Oxford OX1 3RE, UK
| | - Zsofia A Novak
- Sir William Dunn School of Pathology, University of Oxford, Oxford OX1 3RE, UK
| | - Siu-Shing Wong
- Sir William Dunn School of Pathology, University of Oxford, Oxford OX1 3RE, UK
| | - Alain Goriely
- Mathematical Institute, University of Oxford, Oxford OX2 6GG, UK
| | - Michael A Boemo
- Sir William Dunn School of Pathology, University of Oxford, Oxford OX1 3RE, UK.
| | - Jordan W Raff
- Sir William Dunn School of Pathology, University of Oxford, Oxford OX1 3RE, UK.
| |
Collapse
|
227
|
Sullenberger C, Vasquez-Limeta A, Kong D, Loncarek J. With Age Comes Maturity: Biochemical and Structural Transformation of a Human Centriole in the Making. Cells 2020; 9:cells9061429. [PMID: 32526902 PMCID: PMC7349492 DOI: 10.3390/cells9061429] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 05/29/2020] [Accepted: 06/04/2020] [Indexed: 12/14/2022] Open
Abstract
Centrioles are microtubule-based cellular structures present in most human cells that build centrosomes and cilia. Proliferating cells have only two centrosomes and this number is stringently maintained through the temporally and spatially controlled processes of centriole assembly and segregation. The assembly of new centrioles begins in early S phase and ends in the third G1 phase from their initiation. This lengthy process of centriole assembly from their initiation to their maturation is characterized by numerous structural and still poorly understood biochemical changes, which occur in synchrony with the progression of cells through three consecutive cell cycles. As a result, proliferating cells contain three structurally, biochemically, and functionally distinct types of centrioles: procentrioles, daughter centrioles, and mother centrioles. This age difference is critical for proper centrosome and cilia function. Here we discuss the centriole assembly process as it occurs in somatic cycling human cells with a focus on the structural, biochemical, and functional characteristics of centrioles of different ages.
Collapse
|
228
|
Centrosome reduction in newly-generated tetraploid cancer cells obtained by separase depletion. Sci Rep 2020; 10:9152. [PMID: 32499568 PMCID: PMC7272426 DOI: 10.1038/s41598-020-65975-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Accepted: 04/17/2020] [Indexed: 12/29/2022] Open
Abstract
Tetraploidy, a common feature in cancer, results in the presence of extra centrosomes, which has been associated with chromosome instability (CIN) and aneuploidy. Deregulation in the number of centrosomes triggers tumorigenesis. However, how supernumerary centrosomes evolve during the emergence of tetraploid cells remains yet to be elucidated. Here, generating tetraploid isogenic clones in colorectal cancer and in non-transformed cells, we show that near-tetraploid clones exhibit a significant increase in the number of centrosomes. Moreover, we find that centrosome area in near-tetraploids is twice as large as in near-diploids. To evaluate whether centrosome clustering was occurring, we next analysed the number of centrioles revealing centriole amplification. Notwithstanding, more than half of the near-tetraploids maintained in culture do not present centrosome aberrations. To test whether cells progressively lost centrioles after becoming near-tetraploid, we transiently transfected diploid cells with siRNA against ESPL1/Separase, a protease responsible for triggering anaphase, to generate newly near-tetraploid cells. Finally, using this model, we assessed the number of centrioles at different time-points after tetraploidization finding that near-tetraploids rapidly lose centrosomes over time. Taken together, these data demonstrate that although most cells reduce supernumerary centrosomes after tetraploidization, a small fraction retains extra centrioles, potentially resulting in CIN.
Collapse
|
229
|
Arslanhan MD, Gulensoy D, Firat-Karalar EN. A Proximity Mapping Journey into the Biology of the Mammalian Centrosome/Cilium Complex. Cells 2020; 9:E1390. [PMID: 32503249 PMCID: PMC7348975 DOI: 10.3390/cells9061390] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Revised: 05/23/2020] [Accepted: 05/27/2020] [Indexed: 02/07/2023] Open
Abstract
The mammalian centrosome/cilium complex is composed of the centrosome, the primary cilium and the centriolar satellites, which together regulate cell polarity, signaling, proliferation and motility in cells and thereby development and homeostasis in organisms. Accordingly, deregulation of its structure and functions is implicated in various human diseases including cancer, developmental disorders and neurodegenerative diseases. To better understand these disease connections, the molecular underpinnings of the assembly, maintenance and dynamic adaptations of the centrosome/cilium complex need to be uncovered with exquisite detail. Application of proximity-based labeling methods to the centrosome/cilium complex generated spatial and temporal interaction maps for its components and provided key insights into these questions. In this review, we first describe the structure and cell cycle-linked regulation of the centrosome/cilium complex. Next, we explain the inherent biochemical and temporal limitations in probing the structure and function of the centrosome/cilium complex and describe how proximity-based labeling approaches have addressed them. Finally, we explore current insights into the knowledge we gained from the proximity mapping studies as it pertains to centrosome and cilium biogenesis and systematic characterization of the centrosome, cilium and centriolar satellite interactomes.
Collapse
Affiliation(s)
| | | | - Elif Nur Firat-Karalar
- Department of Molecular Biology and Genetics, Koc University, 34450 Istanbul, Turkey; (M.D.A.); (D.G.)
| |
Collapse
|
230
|
Stracker TH, Morrison CG, Gergely F. Molecular causes of primary microcephaly and related diseases: a report from the UNIA Workshop. Chromosoma 2020; 129:115-120. [PMID: 32424716 DOI: 10.1007/s00412-020-00737-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Revised: 05/09/2020] [Accepted: 05/12/2020] [Indexed: 12/30/2022]
Abstract
The International University of Andalucía (UNIA) Current Trends in Biomedicine Workshop on Molecular Causes of Primary Microcephaly and Related Diseases took place in Baeza, Spain, November 18-20, 2019. This meeting brought together scientists from Europe, the USA and China to discuss recent advances in our molecular and genetic understanding of a group of rare neurodevelopmental diseases characterised by primary microcephaly, a condition in which head circumference is smaller than normal at birth. Microcephaly can be caused by inherited mutations that affect key cellular processes, or environmental exposure to radiation or other toxins. It can also result from viral infection, as exemplified by the recent Zika virus outbreak in South America. Here we summarise a number of the scientific advances presented and topics discussed at the meeting.
Collapse
Affiliation(s)
- Travis H Stracker
- Institute for Research in Biomedicine (IRB Barcelona) and Barcelona Institute of Science and Technology, 08028, Barcelona, Spain.
| | - Ciaran G Morrison
- Centre for Chromosome Biology, School of Natural Sciences, National University of Ireland Galway, Biosciences Building, Dangan, Galway, H91 TK33, Ireland
| | - Fanni Gergely
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Robinson Way, Cambridge, CB2 0RE, UK
| |
Collapse
|
231
|
Debec A, Loppin B, Zheng C, Liu X, Megraw TL. The Enigma of Centriole Loss in the 1182-4 Cell Line. Cells 2020; 9:cells9051300. [PMID: 32456186 PMCID: PMC7290863 DOI: 10.3390/cells9051300] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Revised: 05/19/2020] [Accepted: 05/19/2020] [Indexed: 02/07/2023] Open
Abstract
The Drosophila melanogaster cell line 1182-4, which constitutively lacks centrioles, was established many years ago from haploid embryos laid by females homozygous for the maternal haploid (mh) mutation. This was the first clear example of animal cells regularly dividing in the absence of this organelle. However, the cause of the acentriolar nature of the 1182-4 cell line remained unclear and could not be clearly assigned to a particular genetic event. Here, we detail historically the longstanding mystery of the lack of centrioles in this Drosophila cell line. Recent advances, such as the characterization of the mh gene and the genomic analysis of 1182-4 cells, allow now a better understanding of the physiology of these cells. By combining these new data, we propose three reasonable hypotheses of the genesis of this remarkable phenotype.
Collapse
Affiliation(s)
- Alain Debec
- Institute of Ecology and Environmental Sciences, iEES, Sorbonne University, UPEC, CNRS, IRD, INRA, F-75005 Paris, France
- Correspondence: (A.D.); (B.L.); (T.L.M.)
| | - Benjamin Loppin
- Laboratoire de Biologie et de Modélisation de la Cellule—CNRS UMR 5239, École Normale Supérieure de Lyon, University of Lyon, F-69007 Lyon, France
- Correspondence: (A.D.); (B.L.); (T.L.M.)
| | - Chunfeng Zheng
- Department of Biomedical Sciences, College of Medicine, Florida State University, Tallahassee, FL 32306-4300, USA;
| | - Xiuwen Liu
- Department of Computer Science, Florida State University, Tallahassee, FL 32306-4530, USA;
| | - Timothy L. Megraw
- Department of Biomedical Sciences, College of Medicine, Florida State University, Tallahassee, FL 32306-4300, USA;
- Correspondence: (A.D.); (B.L.); (T.L.M.)
| |
Collapse
|
232
|
Sladky VC, Villunger A. Uncovering the PIDDosome and caspase-2 as regulators of organogenesis and cellular differentiation. Cell Death Differ 2020; 27:2037-2047. [PMID: 32415279 PMCID: PMC7308375 DOI: 10.1038/s41418-020-0556-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 04/24/2020] [Accepted: 04/28/2020] [Indexed: 02/08/2023] Open
Abstract
The PIDDosome is a multiprotein complex that drives activation of caspase-2, an endopeptidase originally implicated in apoptosis. Yet, unlike other caspases involved in cell death and inflammation, caspase-2 seems to exert additional versatile functions unrelated to cell death. These emerging roles range from control of transcription factor activity to ploidy surveillance. Thus, caspase-2 and the PIDDosome act as a critical regulatory unit controlling cellular differentiation processes during organogenesis and regeneration. These newly established functions of the PIDDosome and its downstream effector render its components attractive targets for drug-development aiming to prevent fatty liver diseases, neurodegenerative disorders or osteoporosis. ![]()
Collapse
Affiliation(s)
- Valentina C Sladky
- Division of Developmental Immunology, Biocenter, Medical University of Innsbruck, Innsbruck, Austria
| | - Andreas Villunger
- Division of Developmental Immunology, Biocenter, Medical University of Innsbruck, Innsbruck, Austria. .,Ludwig Boltzmann Institute for Rare and Undiagnosed Diseases, 1090, Vienna, Austria. .,CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1090, Vienna, Austria.
| |
Collapse
|
233
|
Requirement of the Cep57-Cep63 Interaction for Proper Cep152 Recruitment and Centriole Duplication. Mol Cell Biol 2020; 40:MCB.00535-19. [PMID: 32152252 DOI: 10.1128/mcb.00535-19] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Accepted: 02/27/2020] [Indexed: 01/27/2023] Open
Abstract
Cep57 has been characterized as a component of a pericentriolar complex containing Cep63 and Cep152. Interestingly, Cep63 and Cep152 self-assemble into a pericentriolar cylindrical architecture, and this event is critical for the orderly recruitment of Plk4, a key regulator of centriole duplication. However, the way in which Cep57 interacts with the Cep63-Cep152 complex and contributes to the structure and function of Cep63-Cep152 self-assembly remains unknown. We demonstrate that Cep57 interacts with Cep63 through N-terminal motifs and associates with Cep152 via Cep63. Three-dimensional structured illumination microscopy (3D-SIM) analyses suggested that the Cep57-Cep63-Cep152 complex is concentrically arranged around a centriole in a Cep57-in and Cep152-out manner. Cep57 mutant cells defective in Cep63 binding exhibited improper Cep63 and Cep152 localization and impaired Sas6 recruitment for procentriole assembly, proving the significance of the Cep57-Cep63 interaction. Intriguingly, Cep63 fused to a microtubule (MT)-binding domain of Cep57 functioned in concert with Cep152 to assemble around stabilized MTs in vitro Thus, Cep57 plays a key role in architecting the Cep63-Cep152 assembly around centriolar MTs and promoting centriole biogenesis. This study may offer a platform to investigate how the organization and function of the pericentriolar architecture are altered by disease-associated mutations found in the Cep57-Cep63-Cep152 complex.
Collapse
|
234
|
Zhao Q, Coughlan KA, Zou MH, Song P. Loss of AMPKalpha1 Triggers Centrosome Amplification via PLK4 Upregulation in Mouse Embryonic Fibroblasts. Int J Mol Sci 2020; 21:ijms21082772. [PMID: 32316320 PMCID: PMC7216113 DOI: 10.3390/ijms21082772] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Revised: 04/15/2020] [Accepted: 04/15/2020] [Indexed: 11/16/2022] Open
Abstract
Recent evidence indicates that activation of adenosine monophosphate-activated protein kinase (AMPK), a highly conserved sensor and modulator of cellular energy and redox, regulates cell mitosis. However, the underlying molecular mechanisms for AMPKα subunit regulation of chromosome segregation remain poorly understood. This study aimed to ascertain if AMPKα1 deletion contributes to chromosome missegregation by elevating Polo-like kinase 4 (PLK4) expression. Centrosome proteins and aneuploidy were monitored in cultured mouse embryonic fibroblasts (MEFs) isolated from wild type (WT, C57BL/6J) or AMPKα1 homozygous deficient (AMPKα1−/−) mice by Western blotting and metaphase chromosome spread. Deletion of AMPKα1, the predominant AMPKα isoform in immortalized MEFs, led to centrosome amplification and chromosome missegregation, as well as the consequent aneuploidy (34–66%) and micronucleus. Furthermore, AMPKα1 null cells exhibited a significant induction of PLK4. Knockdown of nuclear factor kappa B2/p52 ameliorated the PLK4 elevation in AMPKα1-deleted MEFs. Finally, PLK4 inhibition by Centrinone reversed centrosome amplification of AMPKα1-deleted MEFs. Taken together, our results suggest that AMPKα1 plays a fundamental role in the maintenance of chromosomal integrity through the control of p52-mediated transcription of PLK4, a trigger of centriole biogenesis.
Collapse
Affiliation(s)
- Qiang Zhao
- Center for Molecular and Translational Medicine, Georgia State University, Atlanta, GA 30302, USA; (Q.Z.); (M.-H.Z.)
| | | | - Ming-Hui Zou
- Center for Molecular and Translational Medicine, Georgia State University, Atlanta, GA 30302, USA; (Q.Z.); (M.-H.Z.)
| | - Ping Song
- Center for Molecular and Translational Medicine, Georgia State University, Atlanta, GA 30302, USA; (Q.Z.); (M.-H.Z.)
- Correspondence: ; Tel.: +1-404-413-6636
| |
Collapse
|
235
|
Vitre B, Taulet N, Guesdon A, Douanier A, Dosdane A, Cisneros M, Maurin J, Hettinger S, Anguille C, Taschner M, Lorentzen E, Delaval B. IFT proteins interact with HSET to promote supernumerary centrosome clustering in mitosis. EMBO Rep 2020; 21:e49234. [PMID: 32270908 PMCID: PMC7271317 DOI: 10.15252/embr.201949234] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 02/25/2020] [Accepted: 03/12/2020] [Indexed: 11/10/2022] Open
Abstract
Centrosome amplification is a hallmark of cancer, and centrosome clustering is essential for cancer cell survival. The mitotic kinesin HSET is an essential contributor to this process. Recent studies have highlighted novel functions for intraflagellar transport (IFT) proteins in regulating motors and mitotic processes. Here, using siRNA knock‐down of various IFT proteins or AID‐inducible degradation of endogenous IFT88 in combination with small‐molecule inhibition of HSET, we show that IFT proteins together with HSET are required for efficient centrosome clustering. We identify a direct interaction between the kinesin HSET and IFT proteins, and we define how IFT proteins contribute to clustering dynamics during mitosis using high‐resolution live imaging of centrosomes. Finally, we demonstrate the requirement of IFT88 for efficient centrosome clustering in a variety of cancer cell lines naturally harboring supernumerary centrosomes and its importance for cancer cell proliferation. Overall, our data unravel a novel role for the IFT machinery in centrosome clustering during mitosis in cells harboring supernumerary centrosomes.
Collapse
Affiliation(s)
- Benjamin Vitre
- CRBM, University of Montpellier, CNRS, Montpellier, France.,Centrosome, Cilia and Pathologies Lab, Montpellier, France
| | - Nicolas Taulet
- CRBM, University of Montpellier, CNRS, Montpellier, France.,Centrosome, Cilia and Pathologies Lab, Montpellier, France
| | - Audrey Guesdon
- CRBM, University of Montpellier, CNRS, Montpellier, France.,Centrosome, Cilia and Pathologies Lab, Montpellier, France
| | - Audrey Douanier
- CRBM, University of Montpellier, CNRS, Montpellier, France.,Centrosome, Cilia and Pathologies Lab, Montpellier, France
| | - Aurelie Dosdane
- CRBM, University of Montpellier, CNRS, Montpellier, France.,Centrosome, Cilia and Pathologies Lab, Montpellier, France
| | - Melanie Cisneros
- CRBM, University of Montpellier, CNRS, Montpellier, France.,Centrosome, Cilia and Pathologies Lab, Montpellier, France
| | - Justine Maurin
- CRBM, University of Montpellier, CNRS, Montpellier, France.,Centrosome, Cilia and Pathologies Lab, Montpellier, France
| | - Sabrina Hettinger
- CRBM, University of Montpellier, CNRS, Montpellier, France.,Centrosome, Cilia and Pathologies Lab, Montpellier, France
| | - Christelle Anguille
- CRBM, University of Montpellier, CNRS, Montpellier, France.,Centrosome, Cilia and Pathologies Lab, Montpellier, France
| | - Michael Taschner
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | - Esben Lorentzen
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | - Benedicte Delaval
- CRBM, University of Montpellier, CNRS, Montpellier, France.,Centrosome, Cilia and Pathologies Lab, Montpellier, France
| |
Collapse
|
236
|
Hashimoto K, Chinen T, Kitagawa D. Mechanisms of spindle bipolarity establishment in acentrosomal human cells. Mol Cell Oncol 2020; 7:1743899. [PMID: 32391434 PMCID: PMC7199734 DOI: 10.1080/23723556.2020.1743899] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 03/10/2020] [Accepted: 03/12/2020] [Indexed: 06/11/2023]
Abstract
Centrosomes are not absolutely essential for cell division; acentrosomal bipolar spindles can be established in oocytes and centrosome-eliminated somatic cells. However, the detailed mechanisms describing how spindle bipolarity is established without centrosomes are not completely understood. We have recently demonstrated that in acentrosomal human cells, nuclear mitotic apparatus protein (NuMA) assemblies-mediated microtubule asters and EG5 promote spindle bipolarization in early mitosis.
Collapse
Affiliation(s)
- Kaho Hashimoto
- Department of Physiological Chemistry, Graduate School of Pharmaceutical Science, The University of Tokyo, Bunkyo, Tokyo, Japan
| | - Takumi Chinen
- Department of Physiological Chemistry, Graduate School of Pharmaceutical Science, The University of Tokyo, Bunkyo, Tokyo, Japan
| | - Daiju Kitagawa
- Department of Physiological Chemistry, Graduate School of Pharmaceutical Science, The University of Tokyo, Bunkyo, Tokyo, Japan
| |
Collapse
|
237
|
Firat-Karalar EN. Proximity mapping of the microtubule plus-end tracking protein SLAIN2 using the BioID approach. ACTA ACUST UNITED AC 2020; 44:61-72. [PMID: 32256142 PMCID: PMC7129064 DOI: 10.3906/biy-2002-12] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
The centrosome is the main microtubule-organizing center of animal cells, which plays key roles in critical cellular processes ranging from cell division to cellular signaling. Accordingly, defects in the structure and function of centrosomes cause various human diseases such as cancer and primary microcephaly. To elucidate the molecular defects underlying these diseases, the biogenesis and functions of the centrosomes have to be fully understood. An essential step towards addressing these questions is the identification and functional dissection of the full repertoire of centrosome proteins. Here, we used high-resolution imaging and showed that the microtubule plus-end tracking protein SLAIN2 localizes to the pericentriolar material at the proximal end of centrioles. To gain insight into its cellular functions and mechanisms, we applied in vivo proximity-dependent biotin identification to SLAIN2 and generated its proximity interaction map. Gene ontology analysis of the SLAIN2 interactome revealed extensive interactions with centriole duplication, ciliogenesis, and microtubule-associated proteins, including previously characterized and uncharacterized interactions. Collectively, our results define SLAIN2 as a component of pericentriolar material and provide an important resource for future studies aimed at elucidating SLAIN2 functions at the centrosome.
Collapse
Affiliation(s)
- Elif Nur Firat-Karalar
- Department of Molecular Biology and Genetics, Faculty of Science, Koç University, İstanbul Turkey
| |
Collapse
|
238
|
Marthiens V, Basto R. Centrosomes: The good and the bad for brain development. Biol Cell 2020; 112:153-172. [PMID: 32170757 DOI: 10.1111/boc.201900090] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 02/25/2020] [Accepted: 03/01/2020] [Indexed: 12/15/2022]
Abstract
Centrosomes nucleate and organise the microtubule cytoskeleton in animal cells. These membraneless organelles are key structures for tissue organisation, polarity and growth. Centrosome dysfunction, defined as deviation in centrosome numbers and/or structural integrity, has major impact on brain size and functionality, as compared with other tissues of the organism. In this review, we discuss the contribution of centrosomes to brain growth during development. We discuss in particular the impact of centrosome dysfunction in Drosophila and mammalian neural stem cell division and fitness, which ultimately underlie brain growth defects.
Collapse
Affiliation(s)
- Véronique Marthiens
- Biology of Centrosomes and Genetic Instability Laboratory, Institut Curie, PSL Research University, CNRS, UMR144, Paris, 75005, France
| | - Renata Basto
- Biology of Centrosomes and Genetic Instability Laboratory, Institut Curie, PSL Research University, CNRS, UMR144, Paris, 75005, France
| |
Collapse
|
239
|
Riparbelli MG, Persico V, Dallai R, Callaini G. Centrioles and Ciliary Structures during Male Gametogenesis in Hexapoda: Discovery of New Models. Cells 2020; 9:E744. [PMID: 32197383 PMCID: PMC7140630 DOI: 10.3390/cells9030744] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 03/08/2020] [Accepted: 03/10/2020] [Indexed: 12/12/2022] Open
Abstract
Centrioles are-widely conserved barrel-shaped organelles present in most organisms. They are indirectly involved in the organization of the cytoplasmic microtubules both in interphase and during the cell division by recruiting the molecules needed for microtubule nucleation. Moreover, the centrioles are required to assemble cilia and flagella by the direct elongation of their microtubule wall. Due to the importance of the cytoplasmic microtubules in several aspects of the cell life, any defect in centriole structure can lead to cell abnormalities that in humans may result in significant diseases. Many aspects of the centriole dynamics and function have been clarified in the last years, but little attention has been paid to the exceptions in centriole structure that occasionally appeared within the animal kingdom. Here, we focused our attention on non-canonical aspects of centriole architecture within the Hexapoda. The Hexapoda is one of the major animal groups and represents a good laboratory in which to examine the evolution and the organization of the centrioles. Although these findings represent obvious exceptions to the established rules of centriole organization, they may contribute to advance our understanding of the formation and the function of these organelles.
Collapse
Affiliation(s)
- Maria Giovanna Riparbelli
- Department of Life Sciences, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy; (M.G.R.); (V.P.); (R.D.)
| | - Veronica Persico
- Department of Life Sciences, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy; (M.G.R.); (V.P.); (R.D.)
| | - Romano Dallai
- Department of Life Sciences, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy; (M.G.R.); (V.P.); (R.D.)
| | - Giuliano Callaini
- Department of Life Sciences, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy; (M.G.R.); (V.P.); (R.D.)
- Department of Medical Biotechnologies, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy
| |
Collapse
|
240
|
Galletta BJ, Ortega JM, Smith SL, Fagerstrom CJ, Fear JM, Mahadevaraju S, Oliver B, Rusan NM. Sperm Head-Tail Linkage Requires Restriction of Pericentriolar Material to the Proximal Centriole End. Dev Cell 2020; 53:86-101.e7. [PMID: 32169161 DOI: 10.1016/j.devcel.2020.02.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 11/22/2019] [Accepted: 02/07/2020] [Indexed: 01/27/2023]
Abstract
The centriole, or basal body, is the center of attachment between the sperm head and tail. While the distal end of the centriole templates the cilia, the proximal end associates with the nucleus. Using Drosophila, we identify a centriole-centric mechanism that ensures proper proximal end docking to the nucleus. This mechanism relies on the restriction of pericentrin-like protein (PLP) and the pericentriolar material (PCM) to the proximal end of the centriole. PLP is restricted proximally by limiting its mRNA and protein to the earliest stages of centriole elongation. Ectopic positioning of PLP to more distal portions of the centriole is sufficient to redistribute PCM and microtubules along the entire centriole length. This results in erroneous, lateral centriole docking to the nucleus, leading to spermatid decapitation as a result of a failure to form a stable head-tail linkage.
Collapse
Affiliation(s)
- Brian J Galletta
- Cell and Developmental Biology Center, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| | - Jacob M Ortega
- Cell and Developmental Biology Center, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Samantha L Smith
- Cell and Developmental Biology Center, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Carey J Fagerstrom
- Cell and Developmental Biology Center, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Justin M Fear
- Developmental Genomics Section, Laboratory of Cell and Developmental Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Sharvani Mahadevaraju
- Developmental Genomics Section, Laboratory of Cell and Developmental Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Brian Oliver
- Developmental Genomics Section, Laboratory of Cell and Developmental Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Nasser M Rusan
- Cell and Developmental Biology Center, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
241
|
NANOG/NANOGP8 Localizes at the Centrosome and is Spatiotemporally Associated with Centriole Maturation. Cells 2020; 9:cells9030692. [PMID: 32168958 PMCID: PMC7140602 DOI: 10.3390/cells9030692] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 03/06/2020] [Accepted: 03/09/2020] [Indexed: 12/20/2022] Open
Abstract
NANOG is a transcription factor involved in the regulation of pluripotency and stemness. The functional paralog of NANOG, NANOGP8, differs from NANOG in only three amino acids and exhibits similar reprogramming activity. Given the transcriptional regulatory role played by NANOG, the nuclear localization of NANOG/NANOGP8 has primarily been considered to date. In this study, we investigated the intriguing extranuclear localization of NANOG and demonstrated that a substantial pool of NANOG/NANOGP8 is localized at the centrosome. Using double immunofluorescence, the colocalization of NANOG protein with pericentrin was identified by two independent anti-NANOG antibodies among 11 tumor and non-tumor cell lines. The validity of these observations was confirmed by transient expression of GFP-tagged NANOG, which also colocalized with pericentrin. Mass spectrometry of the anti-NANOG immunoprecipitated samples verified the antibody specificity and revealed the expression of both NANOG and NANOGP8, which was further confirmed by real-time PCR. Using cell fractionation, we show that a considerable amount of NANOG protein is present in the cytoplasm of RD and NTERA-2 cells. Importantly, cytoplasmic NANOG was unevenly distributed at the centrosome pair during the cell cycle and colocalized with the distal region of the mother centriole, and its presence was markedly associated with centriole maturation. Along with the finding that the centrosomal localization of NANOG/NANOGP8 was detected in various tumor and non-tumor cell types, these results provide the first evidence suggesting a common centrosome-specific role of NANOG.
Collapse
|
242
|
The tubulin code and its role in controlling microtubule properties and functions. Nat Rev Mol Cell Biol 2020; 21:307-326. [PMID: 32107477 DOI: 10.1038/s41580-020-0214-3] [Citation(s) in RCA: 438] [Impact Index Per Article: 87.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/14/2020] [Indexed: 02/07/2023]
Abstract
Microtubules are core components of the eukaryotic cytoskeleton with essential roles in cell division, shaping, motility and intracellular transport. Despite their functional heterogeneity, microtubules have a highly conserved structure made from almost identical molecular building blocks: the tubulin proteins. Alternative tubulin isotypes and a variety of post-translational modifications control the properties and functions of the microtubule cytoskeleton, a concept known as the 'tubulin code'. Here we review the current understanding of the molecular components of the tubulin code and how they impact microtubule properties and functions. We discuss how tubulin isotypes and post-translational modifications control microtubule behaviour at the molecular level and how this translates into physiological functions at the cellular and organism levels. We then go on to show how fine-tuning of microtubule function by some tubulin modifications can affect homeostasis and how perturbation of this fine-tuning can lead to a range of dysfunctions, many of which are linked to human disease.
Collapse
|
243
|
Targeting centrosome amplification, an Achilles' heel of cancer. Biochem Soc Trans 2020; 47:1209-1222. [PMID: 31506331 PMCID: PMC6824836 DOI: 10.1042/bst20190034] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 08/08/2019] [Accepted: 08/13/2019] [Indexed: 12/12/2022]
Abstract
Due to cell-cycle dysregulation, many cancer cells contain more than the normal compliment of centrosomes, a state referred to as centrosome amplification (CA). CA can drive oncogenic phenotypes and indeed can cause cancer in flies and mammals. However, cells have to actively manage CA, often by centrosome clustering, in order to divide. Thus, CA is also an Achilles' Heel of cancer cells. In recent years, there have been many important studies identifying proteins required for the management of CA and it has been demonstrated that disruption of some of these proteins can cause cancer-specific inhibition of cell growth. For certain targets therapeutically relevant interventions are being investigated, for example, small molecule inhibitors, although none are yet in clinical trials. As the field is now poised to move towards clinically relevant interventions, it is opportune to summarise the key work in targeting CA thus far, with particular emphasis on recent developments where small molecule or other strategies have been proposed. We also highlight the relatively unexplored paradigm of reversing CA, and thus its oncogenic effects, for therapeutic gain.
Collapse
|
244
|
Badarudeen B, Gupta R, Nair SV, Chandrasekharan A, Manna TK. The ubiquitin ligase FBXW7 targets the centriolar assembly protein HsSAS-6 for degradation and thereby regulates centriole duplication. J Biol Chem 2020; 295:4428-4437. [PMID: 32086376 DOI: 10.1074/jbc.ac119.012178] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 02/19/2020] [Indexed: 11/06/2022] Open
Abstract
Formation of a single new centriole from a pre-existing centriole is strictly controlled to maintain correct centrosome number and spindle polarity in cells. However, the mechanisms that govern this process are incompletely understood. Here, using several human cell lines, immunofluorescence and structured illumination microscopy methods, and ubiquitination assays, we show that the E3 ubiquitin ligase F-box and WD repeat domain-containing 7 (FBXW7), a subunit of the SCF ubiquitin ligase, down-regulates spindle assembly 6 homolog (HsSAS-6), a key protein required for procentriole cartwheel assembly, and thereby regulates centriole duplication. We found that FBXW7 abrogation stabilizes HsSAS-6 and increases its recruitment to the mother centriole at multiple sites, leading to supernumerary centrioles. Ultrastructural analyses revealed that FBXW7 is broadly localized on the mother centriole and that its presence is reduced at the site where the HsSAS-6-containing procentriole is formed. This observation suggested that FBXW7 restricts procentriole assembly to a specific site to generate a single new centriole. In contrast, during HsSAS-6 overexpression, FBXW7 strongly associated with HsSAS-6 at the centriole. We also found that SCFFBXW7 interacts with HsSAS-6 and targets it for ubiquitin-mediated degradation. Further, we identified putative phosphodegron sites in HsSAS-6, whose substitutions rendered it insensitive to FBXW7-mediated degradation and control of centriole number. In summary, SCFFBXW7 targets HsSAS-6 for degradation and thereby controls centriole biogenesis by restraining HsSAS-6 recruitment to the mother centriole, a molecular mechanism that controls supernumerary centrioles/centrosomes and the maintenance of bipolar spindles.
Collapse
Affiliation(s)
- Binshad Badarudeen
- School of Biology, Indian Institute of Science Education and Research, Thiruvananthapuram, Vithura, Thiruvananthapuram 695551, Kerala, India
| | - Ria Gupta
- School of Biology, Indian Institute of Science Education and Research, Thiruvananthapuram, Vithura, Thiruvananthapuram 695551, Kerala, India
| | - Sreeja V Nair
- School of Biology, Indian Institute of Science Education and Research, Thiruvananthapuram, Vithura, Thiruvananthapuram 695551, Kerala, India
| | | | - Tapas K Manna
- School of Biology, Indian Institute of Science Education and Research, Thiruvananthapuram, Vithura, Thiruvananthapuram 695551, Kerala, India
| |
Collapse
|
245
|
Phosphorylation of keratin 18 serine 52 regulates mother-daughter centriole engagement and microtubule nucleation by cell cycle-dependent accumulation at the centriole. Histochem Cell Biol 2020; 153:307-321. [PMID: 32078038 DOI: 10.1007/s00418-020-01849-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/23/2020] [Indexed: 12/11/2022]
Abstract
Serine-52 (Ser52) is the major physiologic site of keratin 18 (K18) phosphorylation. Here, we report that serine-52 phosphorylated K18 (phospho-Ser52 K18) accumulated on centrosomes in a cell cycle-dependent manner. Moreover, we found that phospho-Ser52 K18 was located at the proximal end of the mother centriole. Transfection with the K18 Ser52 → Ala (K18 S52A) mutant prevented centriole localization of phospho-Ser52 K18 and resulted in separation of the mother-daughter centrioles. Inhibition of microtubule polymerization led to the disappearance of aggregated phospho-Ser52 K18 on the centrosome; removal of inhibitors resulted in reaccumulation of phospho-Ser52 K18 in microtubule-organizing centers. Transfection with a K18 S52A mutant inhibited microtubule nucleation. These results reveal a cell cycle-dependent change in centrosome localization of phospho-Ser52 k18 and strongly suggest that the phosphorylation status of Ser52 K18 of mother centrioles plays a critical role in maintaining a tight engagement between mother and daughter centrioles and also contributes to microtubule nucleation.
Collapse
|
246
|
Remo A, Li X, Schiebel E, Pancione M. The Centrosome Linker and Its Role in Cancer and Genetic Disorders. Trends Mol Med 2020; 26:380-393. [PMID: 32277932 DOI: 10.1016/j.molmed.2020.01.011] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 11/26/2019] [Accepted: 01/21/2020] [Indexed: 02/07/2023]
Abstract
Centrosome cohesion, the joining of the two centrosomes of a cell, is increasingly appreciated as a major regulator of cell functions such as Golgi organization and cilia positioning. One major element of centrosome cohesion is the centrosome linker that consists of a growing number of proteins. The timely disassembly of the centrosome linker enables centrosomes to separate and assemble a functional bipolar mitotic spindle that is crucial for maintaining genomic integrity. Exciting new findings link centrosome linker defects to cell transformation and genetic disorders. We review recent data on the molecular mechanisms of the assembly and disassembly of the centrosome linker, and discuss how defects in the proper execution of these processes cause DNA damage and genomic instability leading to disease.
Collapse
Affiliation(s)
- Andrea Remo
- Pathology Unit, Mater Salutis Hospital, Azienda Unità Locale Socio Sanitaria (AULSS) 9 'Scaligera', Verona, Italy
| | - Xue Li
- Zentrum für Molekulare Biologie der Universität Heidelberg (ZMBH), Deutsches Krebsforschungszentrum (DKFZ)-ZMBH Allianz, Heidelberg, Germany; Heidelberg Biosciences International Graduate School (HBIGS), Universität Heidelberg, Heidelberg, Germany
| | - Elmar Schiebel
- Zentrum für Molekulare Biologie der Universität Heidelberg (ZMBH), Deutsches Krebsforschungszentrum (DKFZ)-ZMBH Allianz, Heidelberg, Germany.
| | - Massimo Pancione
- Department of Sciences and Technologies, University of Sannio, Benevento, Italy; Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Complutense University of Madrid, Madrid, Spain.
| |
Collapse
|
247
|
Ali H, Braga L, Giacca M. Cardiac regeneration and remodelling of the cardiomyocyte cytoarchitecture. FEBS J 2020; 287:417-438. [PMID: 31743572 DOI: 10.1111/febs.15146] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Revised: 09/27/2019] [Accepted: 11/18/2019] [Indexed: 12/13/2022]
Abstract
Adult mammals are unable to regenerate their hearts after cardiac injury, largely due to the incapacity of cardiomyocytes (CMs) to undergo cell division. However, mammalian embryonic and fetal CMs, similar to CMs from fish and amphibians during their entire life, exhibit robust replicative activity, which stops abruptly after birth and never significantly resumes. Converging evidence indicates that formation of the highly ordered and stable cytoarchitecture of mammalian mature CMs is coupled with loss of their proliferative potential. Here, we review the available information on the role of the cardiac cytoskeleton and sarcomere in the regulation of CM proliferation. The actin cytoskeleton, the intercalated disc, the microtubular network and the dystrophin-glycoprotein complex each sense mechanical cues from the surrounding environment. Furthermore, they participate in the regulation of CM proliferation by impinging on the yes-associated protein/transcriptional co-activator with PDZ-binding motif, β-catenin and myocardin-related transcription factor transcriptional co-activators. Mastering the molecular mechanisms regulating CM proliferation would permit the development of innovative strategies to stimulate cardiac regeneration in adult individuals, a hitherto unachieved yet fundamental therapeutic goal.
Collapse
Affiliation(s)
- Hashim Ali
- British Heart Foundation Centre of Research Excellence, School of Cardiovascular Medicine & Sciences, King's College London, UK.,Molecular Medicine Laboratory, International Centre for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy
| | - Luca Braga
- British Heart Foundation Centre of Research Excellence, School of Cardiovascular Medicine & Sciences, King's College London, UK.,Molecular Medicine Laboratory, International Centre for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy
| | - Mauro Giacca
- British Heart Foundation Centre of Research Excellence, School of Cardiovascular Medicine & Sciences, King's College London, UK.,Molecular Medicine Laboratory, International Centre for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy.,Department of Medical, Surgical and Health Sciences, University of Trieste, Italy
| |
Collapse
|
248
|
Chang SL, Lee SW, Yang SF, Chien CC, Chan TC, Chen TJ, Yang CC, Li CF, Wei YC. Expression and prognostic utility of SSX2IP in patients with nasopharyngeal carcinoma. APMIS 2020; 128:287-297. [PMID: 31837171 DOI: 10.1111/apm.13023] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Accepted: 12/10/2019] [Indexed: 01/20/2023]
Abstract
Cell adhesion affects carcinogenesis, tumor progression, and metastasis. We datamined a published transcriptome (GSE12452) of nasopharyngeal carcinoma (NPC) and identified SSX2IP as a significantly upregulated gene in NPC carcinogenesis among genes associated with cell adhesion (GO:0007155). Consequently, we assessed SSX2IP protein expression and its prognostic significance in 124 patients with NPC using immunohistochemistry and the H-score method. The status of SSX2IP immunoexpression correlated with clinical and pathological characteristics, as well as oncological outcomes. High levels of SSX2IP expression were significantly associated with more advanced primary tumor and TNM stages. Kaplan-Meier and log-rank analyses revealed that high levels of SSX2IP expression, and advanced tumor stage and lymph node metastasis were significantly associated with lower rates of local recurrence-free survival (LRFS), distant metastasis-free survival (DMeFS), and disease-specific (DSS) survival. Multivariate analysis showed that high levels of SSX2IP expression significantly predicted DSS (hazard ratio [HR], 4.290; 95% confidence interval [CI], 2.271-8.102; p < 0.001), DMeFS (HR, 4.159' 95% CI, 2.072-8.345; p < 0.001), and LRFS (HR, 3.007' 95% CI,: 1.418-6.378; p = 0.004). We associated high levels of SSX2IP immunoexpression with aggressive pathological features and worse oncological outcomes, suggesting its potential therapeutic value for patients with NPC.
Collapse
Affiliation(s)
- Shih-Lun Chang
- Department of Otolaryngology, Chi Mei Medical Center, Tainan, Taiwan.,Department of Optometry, Chung Hwa University of Medical Technology, Tainan, Taiwan
| | - Sung-Wei Lee
- Department of Radiation Oncology, Chi Mei Medical Center, Liouying, Taiwan
| | - Sheau-Fang Yang
- Department of Pathology, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan.,Department of Pathology, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chu-Chun Chien
- Department of Pathology, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.,Department of Pathology, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung, Taiwan
| | - Ti-Chun Chan
- Department of Medical Research, Chi Mei Medical Center, Tainan, Taiwan
| | - Tzu-Ju Chen
- Department of Pathology, Chi Mei Medical Center, Tainan, Taiwan.,Department of Optometry, Chung Hwa University of Medical Technology, Tainan, Taiwan.,Institute of Biomedical Science, National Sun Yat-sen University, Kaohsiung, Taiwan
| | - Ching-Chieh Yang
- Department of Radiation Oncology, Chi Mei Medical Center, Tainan, Taiwan.,Department of Pharmacy, Chia-Nan University of Pharmacy and Science, Tainan, Taiwan
| | - Chien-Feng Li
- Department of Pathology, Chi Mei Medical Center, Tainan, Taiwan.,National Institute of Cancer Research, National Health Research Institute, Tainan, Taiwan.,Department of Biotechnology, Southern Taiwan University of Science and Technology, Tainan, Taiwan
| | - Yu-Ching Wei
- Department of Pathology, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.,Department of Pathology, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung, Taiwan
| |
Collapse
|
249
|
Ledur PF, Karmirian K, Pedrosa CDSG, Souza LRQ, Assis-de-Lemos G, Martins TM, Ferreira JDCCG, de Azevedo Reis GF, Silva ES, Silva D, Salerno JA, Ornelas IM, Devalle S, Madeiro da Costa RF, Goto-Silva L, Higa LM, Melo A, Tanuri A, Chimelli L, Murata MM, Garcez PP, Filippi-Chiela EC, Galina A, Borges HL, Rehen SK. Zika virus infection leads to mitochondrial failure, oxidative stress and DNA damage in human iPSC-derived astrocytes. Sci Rep 2020; 10:1218. [PMID: 31988337 PMCID: PMC6985105 DOI: 10.1038/s41598-020-57914-x] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Accepted: 01/02/2020] [Indexed: 12/14/2022] Open
Abstract
Zika virus (ZIKV) has been extensively studied since it was linked to congenital malformations, and recent research has revealed that astrocytes are targets of ZIKV. However, the consequences of ZIKV infection, especially to this cell type, remain largely unknown, particularly considering integrative studies aiming to understand the crosstalk among key cellular mechanisms and fates involved in the neurotoxicity of the virus. Here, the consequences of ZIKV infection in iPSC-derived astrocytes are presented. Our results show ROS imbalance, mitochondrial defects and DNA breakage, which have been previously linked to neurological disorders. We have also detected glial reactivity, also present in mice and in post-mortem brains from infected neonates from the Northeast of Brazil. Given the role of glia in the developing brain, these findings may help to explain the observed effects in congenital Zika syndrome related to neuronal loss and motor deficit.
Collapse
Affiliation(s)
| | - Karina Karmirian
- D'Or Institute for Research and Education, Rio de Janeiro, Brazil
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, RJ, Brazil
| | | | | | - Gabriela Assis-de-Lemos
- Institute of Medical Biochemistry Leopoldo De Meis, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Thiago Martino Martins
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, RJ, Brazil
| | | | - Gabriel Ferreira de Azevedo Reis
- Insitute of Biology, Department of Biophysics and Biometrics, State University of Rio de Janeiro (UERJ), Rio de Janeiro, RJ, Brazil
| | - Eduardo Santos Silva
- Insitute of Biology, Department of Biophysics and Biometrics, State University of Rio de Janeiro (UERJ), Rio de Janeiro, RJ, Brazil
| | - Débora Silva
- Laboratory of Neuropathology, State Institute of Brain Paulo Niemeyer, Rio de Janeiro, RJ, Brazil
| | - José Alexandre Salerno
- D'Or Institute for Research and Education, Rio de Janeiro, Brazil
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, RJ, Brazil
| | | | - Sylvie Devalle
- D'Or Institute for Research and Education, Rio de Janeiro, Brazil
| | | | - Livia Goto-Silva
- D'Or Institute for Research and Education, Rio de Janeiro, Brazil
| | - Luiza Mendonça Higa
- Institute of Biology, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, RJ, Brazil
| | - Adriana Melo
- Research Institute Prof. Joaquim Amorim Neto (IPESQ), Campina Grande, PB, Brazil
| | - Amilcar Tanuri
- Institute of Biology, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, RJ, Brazil
| | - Leila Chimelli
- Laboratory of Neuropathology, State Institute of Brain Paulo Niemeyer, Rio de Janeiro, RJ, Brazil
| | - Marcos Massao Murata
- Insitute of Biology, Department of Biophysics and Biometrics, State University of Rio de Janeiro (UERJ), Rio de Janeiro, RJ, Brazil
| | - Patrícia Pestana Garcez
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, RJ, Brazil
| | | | - Antonio Galina
- Institute of Medical Biochemistry Leopoldo De Meis, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Helena Lobo Borges
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, RJ, Brazil
| | - Stevens Kastrup Rehen
- D'Or Institute for Research and Education, Rio de Janeiro, Brazil.
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, RJ, Brazil.
| |
Collapse
|
250
|
E2F-Family Members Engage the PIDDosome to Limit Hepatocyte Ploidy in Liver Development and Regeneration. Dev Cell 2020; 52:335-349.e7. [PMID: 31983631 DOI: 10.1016/j.devcel.2019.12.016] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 10/27/2019] [Accepted: 12/24/2019] [Indexed: 01/12/2023]
Abstract
E2F transcription factors control the cytokinesis machinery and thereby ploidy in hepatocytes. If or how these proteins limit proliferation of polyploid cells with extra centrosomes remains unknown. Here, we show that the PIDDosome, a signaling platform essential for caspase-2-activation, limits hepatocyte ploidy and is instructed by the E2F network to control p53 in the developing as well as regenerating liver. Casp2 and Pidd1 act as direct transcriptional targets of E2F1 and its antagonists, E2F7 and E2F8, that together co-regulate PIDDosome expression during juvenile liver growth and regeneration. Of note, whereas hepatocyte aneuploidy correlates with the basal ploidy state, the degree of aneuploidy itself is not limited by PIDDosome-dependent p53 activation. Finally, we provide evidence that the same signaling network is engaged to control ploidy in the human liver after resection. Our study defines the PIDDosome as a primary target to manipulate hepatocyte ploidy and proliferation rates in the regenerating liver.
Collapse
|