1
|
Serizay J, Khoury Damaa M, Boudjema AR, Balagué R, Faucourt M, Delgehyr N, Noûs C, Zaragosi LE, Barbry P, Spassky N, Koszul R, Meunier A. Cyclin switch tailors a cell cycle variant to orchestrate multiciliogenesis. Cell Rep 2024:115103. [PMID: 39740664 DOI: 10.1016/j.celrep.2024.115103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 11/19/2024] [Accepted: 12/03/2024] [Indexed: 01/02/2025] Open
Abstract
Meiosis, endoreplication, and asynthetic fissions are variations of the canonical cell cycle where either replication or mitotic divisions are muted. Here, we identify a cell cycle variantconserved across organs and mammals, where both replication and mitosis are muted, and that orchestrates the differentiation of post-mitotic progenitors into multiciliated cells (MCCs). MCC progenitors reactivate most of the cell cycle transcriptional program but replace the temporal expression of cyclins E2 and A2 with non-canonical cyclins O and A1. In addition, the primary APC/C inhibitor Emi1 is silenced. Re-expressing cyclins E2 and A2 and/or Emi1 can induce partial replication or mitosis. This shows that a cell can co-opt the cell cycle genetic program and regulate only certain elements to qualitatively and quantitatively divert CDK activity toward differentiation rather than division. We propose this cell cycle variant to exploit the existence of a cytoplasmic-or centriolar-CDK threshold lower than the S-phase threshold.
Collapse
Affiliation(s)
- Jacques Serizay
- Institut de Biologie de l'ENS (IBENS), CNRS, INSERM, École Normale Supérieure, PSL Research University, Paris, France; Institut Pasteur, CNRS UMR3525, Université Paris Cité, Unité Régulation Spatiale des Génomes, Paris, France.
| | - Michella Khoury Damaa
- Institut de Biologie de l'ENS (IBENS), CNRS, INSERM, École Normale Supérieure, PSL Research University, Paris, France
| | - Amélie-Rose Boudjema
- Institut de Biologie de l'ENS (IBENS), CNRS, INSERM, École Normale Supérieure, PSL Research University, Paris, France
| | - Rémi Balagué
- Institut de Biologie de l'ENS (IBENS), CNRS, INSERM, École Normale Supérieure, PSL Research University, Paris, France
| | - Marion Faucourt
- Institut de Biologie de l'ENS (IBENS), CNRS, INSERM, École Normale Supérieure, PSL Research University, Paris, France
| | - Nathalie Delgehyr
- Institut de Biologie de l'ENS (IBENS), CNRS, INSERM, École Normale Supérieure, PSL Research University, Paris, France
| | - Camille Noûs
- Cogitamus Laboratory, PSL University, Paris, France
| | - Laure-Emmanuelle Zaragosi
- Université Côte d'Azur, CNRS, Institut de Pharmacologie Moléculaire et Cellulaire, 06560 Sophia Antipolis, France
| | - Pascal Barbry
- Université Côte d'Azur, CNRS, Institut de Pharmacologie Moléculaire et Cellulaire, 06560 Sophia Antipolis, France
| | - Nathalie Spassky
- Institut de Biologie de l'ENS (IBENS), CNRS, INSERM, École Normale Supérieure, PSL Research University, Paris, France
| | - Romain Koszul
- Institut Pasteur, CNRS UMR3525, Université Paris Cité, Unité Régulation Spatiale des Génomes, Paris, France
| | - Alice Meunier
- Institut de Biologie de l'ENS (IBENS), CNRS, INSERM, École Normale Supérieure, PSL Research University, Paris, France.
| |
Collapse
|
2
|
Joo YK, Ramirez C, Kabeche L. A TRilogy of ATR's Non-Canonical Roles Throughout the Cell Cycle and Its Relation to Cancer. Cancers (Basel) 2024; 16:3536. [PMID: 39456630 PMCID: PMC11506335 DOI: 10.3390/cancers16203536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 10/12/2024] [Accepted: 10/17/2024] [Indexed: 10/28/2024] Open
Abstract
Ataxia Telangiectasia and Rad3-related protein (ATR) is an apical kinase of the DNA Damage Response (DDR) pathway responsible for detecting and resolving damaged DNA. Because cancer cells depend heavily on the DNA damage checkpoint for their unchecked proliferation and propagation, ATR has gained enormous popularity as a cancer therapy target in recent decades. Yet, ATR inhibitors have not been the silver bullets as anticipated, with clinical trials demonstrating toxicity and mixed efficacy. To investigate whether the toxicity and mixed efficacy of ATR inhibitors arise from their off-target effects related to ATR's multiple roles within and outside the DDR pathway, we have analyzed recently published studies on ATR's non-canonical roles. Recent studies have elucidated that ATR plays a wide role throughout the cell cycle that is separate from its function in the DDR. This includes maintaining nuclear membrane integrity, detecting mechanical forces, and promoting faithful chromosome segregation during mitosis. In this review, we summarize the canonical, DDR-related roles of ATR and also focus on the non-canonical, multifaceted roles of ATR throughout the cell cycle and their clinical relevance. Through this summary, we also address the need for re-assessing clinical strategies targeting ATR as a cancer therapy based on these newly discovered roles for ATR.
Collapse
Affiliation(s)
- Yoon Ki Joo
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT 06511, USA
- Yale Cancer Biology Institute, Yale University, West Haven, CT 06516, USA
| | - Carlos Ramirez
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT 06511, USA
- Yale Cancer Biology Institute, Yale University, West Haven, CT 06516, USA
| | - Lilian Kabeche
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT 06511, USA
- Yale Cancer Biology Institute, Yale University, West Haven, CT 06516, USA
| |
Collapse
|
3
|
Vicars H, Karg T, Mills A, Sullivan W. Acentric chromosome congression and alignment on the metaphase plate via kinetochore-independent forces in Drosophila. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.11.14.567057. [PMID: 38798431 PMCID: PMC11118298 DOI: 10.1101/2023.11.14.567057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Chromosome congression and alignment on the metaphase plate involves lateral and microtubule plus-end interactions with the kinetochore. Here we take advantage of our ability to efficiently generate a GFP-marked acentric X chromosome fragment in Drosophila neuroblasts to identify forces acting on chromosome arms that drive congression and alignment. We find acentrics efficiently align on the metaphase plate, often more rapidly than kinetochore-bearing chromosomes. Unlike intact chromosomes, the paired sister acentrics oscillate as they move to and reside on the metaphase plate in a plane distinct and significantly further from the main mass of intact chromosomes. Consequently, at anaphase onset acentrics are oriented either parallel or perpendicular to the spindle. Parallel-oriented sisters separate by sliding while those oriented perpendicularly separate via unzipping. This oscillation, together with the fact that in monopolar spindles acentrics are rapidly shunted away from the poles, indicates that distributed plus-end directed forces are primarily responsible for acentric migration. This conclusion is supported by the observation that reduction of EB1 preferentially disrupts acentric alignment. In addition, reduction of Klp3a activity, a gene required for the establishment of pole-to-pole microtubules, preferentially disrupts acentric alignment. Taken together these studies suggest that plus-end forces mediated by the outer pole-to-pole microtubules are primarily responsible for acentric metaphase alignment. Surprisingly, we find that a small fraction of sister acentrics are anti-parallel aligned indicating that the kinetochore is required to ensure parallel alignment of sister chromatids. Finally, we find induction of acentric chromosome fragments results in a global reorganization of the congressed chromosomes into a torus configuration. Article Summary The kinetochore serves as a site for attaching microtubules and allows for successful alignment, separation, and segregation of replicated sister chromosomes during cell division. However, previous studies have revealed that sister chromosomes without kinetochores (acentrics) often align to the metaphase plate, undergo separation and segregation, and are properly transmitted to daughter cells. In this study, we discuss the forces acting on chromosomes, independent of the kinetochore, underlying their successful alignment in early mitosis.
Collapse
|
4
|
Athwal H, Kochiyanil A, Bhat V, Allan AL, Parsyan A. Centrosomes and associated proteins in pathogenesis and treatment of breast cancer. Front Oncol 2024; 14:1370565. [PMID: 38606093 PMCID: PMC11007099 DOI: 10.3389/fonc.2024.1370565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Accepted: 03/04/2024] [Indexed: 04/13/2024] Open
Abstract
Breast cancer is the most prevalent malignancy among women worldwide. Despite significant advances in treatment, it remains one of the leading causes of female mortality. The inability to effectively treat advanced and/or treatment-resistant breast cancer demonstrates the need to develop novel treatment strategies and targeted therapies. Centrosomes and their associated proteins have been shown to play key roles in the pathogenesis of breast cancer and thus represent promising targets for drug and biomarker development. Centrosomes are fundamental cellular structures in the mammalian cell that are responsible for error-free execution of cell division. Centrosome amplification and aberrant expression of its associated proteins such as Polo-like kinases (PLKs), Aurora kinases (AURKs) and Cyclin-dependent kinases (CDKs) have been observed in various cancers, including breast cancer. These aberrations in breast cancer are thought to cause improper chromosomal segregation during mitosis, leading to chromosomal instability and uncontrolled cell division, allowing cancer cells to acquire new genetic changes that result in evasion of cell death and the promotion of tumor formation. Various chemical compounds developed against PLKs and AURKs have shown meaningful antitumorigenic effects in breast cancer cells in vitro and in vivo. The mechanism of action of these inhibitors is likely related to exacerbation of numerical genomic instability, such as aneuploidy or polyploidy. Furthermore, growing evidence demonstrates enhanced antitumorigenic effects when inhibitors specific to centrosome-associated proteins are used in combination with either radiation or chemotherapy drugs in breast cancer. This review focuses on the current knowledge regarding the roles of centrosome and centrosome-associated proteins in breast cancer pathogenesis and their utility as novel targets for breast cancer treatment.
Collapse
Affiliation(s)
- Harjot Athwal
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| | - Arpitha Kochiyanil
- Faculty of Science, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| | - Vasudeva Bhat
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
- London Regional Cancer Program, London Health Sciences Centre, Lawson Health Research Institute, London, ON, Canada
| | - Alison L. Allan
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
- London Regional Cancer Program, London Health Sciences Centre, Lawson Health Research Institute, London, ON, Canada
- Department of Oncology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| | - Armen Parsyan
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
- London Regional Cancer Program, London Health Sciences Centre, Lawson Health Research Institute, London, ON, Canada
- Department of Oncology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
- Division of General Surgery, Department of Surgery, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
- Department of Surgery, St. Joseph’s Health Care London and London Health Sciences Centre, London, ON, Canada
| |
Collapse
|
5
|
Schindler-Johnson M, Petridou NI. Collective effects of cell cleavage dynamics. Front Cell Dev Biol 2024; 12:1358971. [PMID: 38559810 PMCID: PMC10978805 DOI: 10.3389/fcell.2024.1358971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 03/05/2024] [Indexed: 04/04/2024] Open
Abstract
A conserved process of early embryonic development in metazoans is the reductive cell divisions following oocyte fertilization, termed cell cleavages. Cell cleavage cycles usually start synchronously, lengthen differentially between the embryonic cells becoming asynchronous, and cease before major morphogenetic events, such as germ layer formation and gastrulation. Despite exhibiting species-specific characteristics, the regulation of cell cleavage dynamics comes down to common controllers acting mostly at the single cell/nucleus level, such as nucleus-to-cytoplasmic ratio and zygotic genome activation. Remarkably, recent work has linked cell cleavage dynamics to the emergence of collective behavior during embryogenesis, including pattern formation and changes in embryo-scale mechanics, raising the question how single-cell controllers coordinate embryo-scale processes. In this review, we summarize studies across species where an association between cell cleavages and collective behavior was made, discuss the underlying mechanisms, and propose that cell-to-cell variability in cell cleavage dynamics can serve as a mechanism of long-range coordination in developing embryos.
Collapse
Affiliation(s)
- Magdalena Schindler-Johnson
- Developmental Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Nicoletta I. Petridou
- Developmental Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| |
Collapse
|
6
|
Wang Q, Bode AM, Zhang T. Targeting CDK1 in cancer: mechanisms and implications. NPJ Precis Oncol 2023; 7:58. [PMID: 37311884 DOI: 10.1038/s41698-023-00407-7] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 05/25/2023] [Indexed: 06/15/2023] Open
Abstract
Cyclin dependent kinases (CDKs) are serine/threonine kinases that are proposed as promising candidate targets for cancer treatment. These proteins complexed with cyclins play a critical role in cell cycle progression. Most CDKs demonstrate substantially higher expression in cancer tissues compared with normal tissues and, according to the TCGA database, correlate with survival rate in multiple cancer types. Deregulation of CDK1 has been shown to be closely associated with tumorigenesis. CDK1 activation plays a critical role in a wide range of cancer types; and CDK1 phosphorylation of its many substrates greatly influences their function in tumorigenesis. Enrichment of CDK1 interacting proteins with Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis was conducted to demonstrate that the associated proteins participate in multiple oncogenic pathways. This abundance of evidence clearly supports CDK1 as a promising target for cancer therapy. A number of small molecules targeting CDK1 or multiple CDKs have been developed and evaluated in preclinical studies. Notably, some of these small molecules have also been subjected to human clinical trials. This review evaluates the mechanisms and implications of targeting CDK1 in tumorigenesis and cancer therapy.
Collapse
Affiliation(s)
- Qiushi Wang
- The Hormel Institute, University of Minnesota, 801 16th Ave NE, Austin, MN, 55912, USA
| | - Ann M Bode
- The Hormel Institute, University of Minnesota, 801 16th Ave NE, Austin, MN, 55912, USA.
| | - Tianshun Zhang
- The Hormel Institute, University of Minnesota, 801 16th Ave NE, Austin, MN, 55912, USA.
| |
Collapse
|
7
|
Abstract
Understanding the mechanisms of embryonic cell cycles is a central goal of developmental biology, as the regulation of the cell cycle must be closely coordinated with other events during early embryogenesis. Quantitative imaging approaches have recently begun to reveal how the cell cycle oscillator is controlled in space and time, and how it is integrated with mechanical signals to drive morphogenesis. Here, we discuss how the Drosophila embryo has served as an excellent model for addressing the molecular and physical mechanisms of embryonic cell cycles, with comparisons to other model systems to highlight conserved and species-specific mechanisms. We describe how the rapid cleavage divisions characteristic of most metazoan embryos require chemical waves and cytoplasmic flows to coordinate morphogenesis across the large expanse of the embryo. We also outline how, in the late cleavage divisions, the cell cycle is inter-regulated with the activation of gene expression to ensure a reliable maternal-to-zygotic transition. Finally, we discuss how precise transcriptional regulation of the timing of mitosis ensures that tissue morphogenesis and cell proliferation are tightly controlled during gastrulation.
Collapse
Affiliation(s)
| | - Stefano Di Talia
- Department of Cell Biology, Duke University Medical Center, Durham, NC 27705, USA
| |
Collapse
|
8
|
Modeling the role for nuclear import dynamics in the early embryonic cell cycle. Biophys J 2021; 120:4277-4286. [PMID: 34022240 DOI: 10.1016/j.bpj.2021.05.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 04/22/2021] [Accepted: 05/06/2021] [Indexed: 11/21/2022] Open
Abstract
Nuclear composition determines nuclear function. The early embryos of many species begin life with large pools of maternally provided components that become rapidly imported into an increasing number of nuclei as the cells undergo repeated cleavage divisions. Because early cell cycles are too fast for nuclei to achieve steady-state nucleocytoplasmic partitioning, the composition of cleavage stage nuclei is likely dominated by nuclear import. The end of the rapid cleavage stage and onset of major zygotic transcription, known as the mid-blastula transition (MBT), is controlled by the ratio of nuclei/cytoplasm, indicating that changes in nuclear composition likely mediate MBT timing. Here, we explore how different nuclear import regimes can affect protein accumulation in the nucleus in the early Drosophila embryo. We find that nuclear import differs dramatically for a general nuclear cargo (NLS (nuclear localization signal)-mRFP) and a proposed MBT regulator (histone H3). We show that nuclear import rates of NLS-mRFP in a given nucleus remain relatively unchanged throughout the cleavage cycles, whereas those of H3 halve with each cycle. We model these two distinct modes of nuclear import as "nucleus-limited" and "import-limited" and examine how the two different modes can contribute to different protein accumulation dynamics. Finally, we incorporate these distinct modes of nuclear import into a model for cell-cycle regulation at the MBT and find that the import-limited H3 dynamics contribute to increased robustness and allow for stepwise cell-cycle slowing at the MBT.
Collapse
|
9
|
Vicars H, Karg T, Warecki B, Bast I, Sullivan W. Kinetochore-independent mechanisms of sister chromosome separation. PLoS Genet 2021; 17:e1009304. [PMID: 33513180 PMCID: PMC7886193 DOI: 10.1371/journal.pgen.1009304] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 02/16/2021] [Accepted: 12/08/2020] [Indexed: 11/19/2022] Open
Abstract
Although kinetochores normally play a key role in sister chromatid separation and segregation, chromosome fragments lacking kinetochores (acentrics) can in some cases separate and segregate successfully. In Drosophila neuroblasts, acentric chromosomes undergo delayed, but otherwise normal sister separation, revealing the existence of kinetochore- independent mechanisms driving sister chromosome separation. Bulk cohesin removal from the acentric is not delayed, suggesting factors other than cohesin are responsible for the delay in acentric sister separation. In contrast to intact kinetochore-bearing chromosomes, we discovered that acentrics align parallel as well as perpendicular to the mitotic spindle. In addition, sister acentrics undergo unconventional patterns of separation. For example, rather than the simultaneous separation of sisters, acentrics oriented parallel to the spindle often slide past one another toward opposing poles. To identify the mechanisms driving acentric separation, we screened 117 RNAi gene knockdowns for synthetic lethality with acentric chromosome fragments. In addition to well-established DNA repair and checkpoint mutants, this candidate screen identified synthetic lethality with X-chromosome-derived acentric fragments in knockdowns of Greatwall (cell cycle kinase), EB1 (microtubule plus-end tracking protein), and Map205 (microtubule-stabilizing protein). Additional image-based screening revealed that reductions in Topoisomerase II levels disrupted sister acentric separation. Intriguingly, live imaging revealed that knockdowns of EB1, Map205, and Greatwall preferentially disrupted the sliding mode of sister acentric separation. Based on our analysis of EB1 localization and knockdown phenotypes, we propose that in the absence of a kinetochore, microtubule plus-end dynamics provide the force to resolve DNA catenations required for sister separation.
Collapse
Affiliation(s)
- Hannah Vicars
- Department of Molecular, Cell and Developmental Biology, University of California, Santa Cruz, Santa Cruz, California, United States of America
| | - Travis Karg
- Department of Molecular, Cell and Developmental Biology, University of California, Santa Cruz, Santa Cruz, California, United States of America
| | - Brandt Warecki
- Department of Molecular, Cell and Developmental Biology, University of California, Santa Cruz, Santa Cruz, California, United States of America
| | - Ian Bast
- Department of Molecular, Cell and Developmental Biology, University of California, Santa Cruz, Santa Cruz, California, United States of America
| | - William Sullivan
- Department of Molecular, Cell and Developmental Biology, University of California, Santa Cruz, Santa Cruz, California, United States of America
| |
Collapse
|
10
|
Chari S, Wilky H, Govindan J, Amodeo AA. Histone concentration regulates the cell cycle and transcription in early development. Development 2019; 146:dev.177402. [PMID: 31511251 DOI: 10.1242/dev.177402] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 08/28/2019] [Indexed: 12/12/2022]
Abstract
The early embryos of many animals, including flies, fish and frogs, have unusually rapid cell cycles and delayed onset of transcription. These divisions are dependent on maternally supplied RNAs and proteins including histones. Previous work suggests that the pool size of maternally provided histones can alter the timing of zygotic genome activation (ZGA) in frogs and fish. Here, we examine the effects of under- and overexpression of maternal histones in Drosophila embryogenesis. Decreasing histone concentration advances zygotic transcription, cell cycle elongation, Chk1 activation and gastrulation. Conversely, increasing histone concentration delays transcription and results in an additional nuclear cycle before gastrulation. Numerous zygotic transcripts are sensitive to histone concentration, and the promoters of histone-sensitive genes are associated with specific chromatin features linked to increased histone turnover. These include enrichment of the pioneer transcription factor Zelda, and lack of SIN3A and associated histone deacetylases. Our findings uncover a crucial regulatory role for histone concentrations in ZGA of Drosophila.
Collapse
Affiliation(s)
- Sudarshan Chari
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544, USA
| | - Henry Wilky
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544, USA
| | - Jayalakshmi Govindan
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544, USA
| | - Amanda A Amodeo
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544, USA
| |
Collapse
|
11
|
Joukov V, De Nicolo A. The Centrosome and the Primary Cilium: The Yin and Yang of a Hybrid Organelle. Cells 2019; 8:E701. [PMID: 31295970 PMCID: PMC6678760 DOI: 10.3390/cells8070701] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 07/04/2019] [Accepted: 07/06/2019] [Indexed: 12/27/2022] Open
Abstract
Centrosomes and primary cilia are usually considered as distinct organelles, although both are assembled with the same evolutionary conserved, microtubule-based templates, the centrioles. Centrosomes serve as major microtubule- and actin cytoskeleton-organizing centers and are involved in a variety of intracellular processes, whereas primary cilia receive and transduce environmental signals to elicit cellular and organismal responses. Understanding the functional relationship between centrosomes and primary cilia is important because defects in both structures have been implicated in various diseases, including cancer. Here, we discuss evidence that the animal centrosome evolved, with the transition to complex multicellularity, as a hybrid organelle comprised of the two distinct, but intertwined, structural-functional modules: the centriole/primary cilium module and the pericentriolar material/centrosome module. The evolution of the former module may have been caused by the expanding cellular diversification and intercommunication, whereas that of the latter module may have been driven by the increasing complexity of mitosis and the requirement for maintaining cell polarity, individuation, and adhesion. Through its unique ability to serve both as a plasma membrane-associated primary cilium organizer and a juxtanuclear microtubule-organizing center, the animal centrosome has become an ideal integrator of extracellular and intracellular signals with the cytoskeleton and a switch between the non-cell autonomous and the cell-autonomous signaling modes. In light of this hypothesis, we discuss centrosome dynamics during cell proliferation, migration, and differentiation and propose a model of centrosome-driven microtubule assembly in mitotic and interphase cells. In addition, we outline the evolutionary benefits of the animal centrosome and highlight the hierarchy and modularity of the centrosome biogenesis networks.
Collapse
Affiliation(s)
- Vladimir Joukov
- N.N. Petrov National Medical Research Center of Oncology, 197758 Saint-Petersburg, Russia.
| | | |
Collapse
|
12
|
del Castillo U, Gnazzo MM, Turpin CGS, Nguyen KCQ, Semaya E, Lam Y, de Cruz MA, Bembenek JN, Hall DH, Riggs B, Gelfand VI, Skop AR. Conserved role for Ataxin-2 in mediating endoplasmic reticulum dynamics. Traffic 2019; 20:436-447. [PMID: 30989774 PMCID: PMC6553494 DOI: 10.1111/tra.12647] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Revised: 04/04/2019] [Accepted: 04/05/2019] [Indexed: 12/17/2022]
Abstract
Ataxin-2, a conserved RNA-binding protein, is implicated in the late-onset neurodegenerative disease Spinocerebellar ataxia type-2 (SCA2). SCA2 is characterized by shrunken dendritic arbors and torpedo-like axons within the Purkinje neurons of the cerebellum. Torpedo-like axons have been described to contain displaced endoplasmic reticulum (ER) in the periphery of the cell; however, the role of Ataxin-2 in mediating ER function in SCA2 is unclear. We utilized the Caenorhabditis elegans and Drosophila homologs of Ataxin-2 (ATX-2 and DAtx2, respectively) to determine the role of Ataxin-2 in ER function and dynamics in embryos and neurons. Loss of ATX-2 and DAtx2 resulted in collapse of the ER in dividing embryonic cells and germline, and ultrastructure analysis revealed unique spherical stacks of ER in mature oocytes and fragmented and truncated ER tubules in the embryo. ATX-2 and DAtx2 reside in puncta adjacent to the ER in both C. elegans and Drosophila embryos. Lastly, depletion of DAtx2 in cultured Drosophila neurons recapitulated the shrunken dendritic arbor phenotype of SCA2. ER morphology and dynamics were severely disrupted in these neurons. Taken together, we provide evidence that Ataxin-2 plays an evolutionary conserved role in ER dynamics and morphology in C. elegans and Drosophila embryos during development and in fly neurons, suggesting a possible SCA2 disease mechanism.
Collapse
Affiliation(s)
- Urko del Castillo
- Department of Cell and Molecular Biology, Feinberg School of Medicine, Northwestern University, 303 E Chicago Ave, Chicago, IL 60611
| | - Megan M. Gnazzo
- Laboratory of Genetics, University of Wisconsin-Madison, Madison, WI 53706
| | - Christopher G. Sorensen Turpin
- Department of Biochemistry and Cellular and Molecular Biology, University of Tennessee-Knoxville, Knoxville, Tennessee 37996
| | - Ken C. Q. Nguyen
- Center for C. elegans Anatomy, Albert Einstein College of Medicine, Bronx New York, NY 10461
| | - Emily Semaya
- Center for C. elegans Anatomy, Albert Einstein College of Medicine, Bronx New York, NY 10461
| | - Yuwan Lam
- Department of Biology, San Francisco State University, San Francisco, CA 94132
| | - Matthew A. de Cruz
- Department of Biology, San Francisco State University, San Francisco, CA 94132
| | - Joshua N. Bembenek
- Department of Biochemistry and Cellular and Molecular Biology, University of Tennessee-Knoxville, Knoxville, Tennessee 37996
| | - David H. Hall
- Center for C. elegans Anatomy, Albert Einstein College of Medicine, Bronx New York, NY 10461
| | - Blake Riggs
- Department of Biology, San Francisco State University, San Francisco, CA 94132
| | - Vladimir I. Gelfand
- Department of Cell and Molecular Biology, Feinberg School of Medicine, Northwestern University, 303 E Chicago Ave, Chicago, IL 60611
| | - Ahna R. Skop
- Laboratory of Genetics, University of Wisconsin-Madison, Madison, WI 53706
| |
Collapse
|
13
|
Autophagy maintains stem cells and intestinal homeostasis in Drosophila. Sci Rep 2018; 8:4644. [PMID: 29545557 PMCID: PMC5854693 DOI: 10.1038/s41598-018-23065-3] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Accepted: 03/05/2018] [Indexed: 12/12/2022] Open
Abstract
Intestinal homeostasis is maintained by tightly controlled proliferation and differentiation of tissue-resident multipotent stem cells during aging and regeneration, which ensures organismal adaptation. Here we show that autophagy is required in Drosophila intestinal stem cells to sustain proliferation, and preserves the stem cell pool. Autophagy-deficient stem cells show elevated DNA damage and cell cycle arrest during aging, and are frequently eliminated via JNK-mediated apoptosis. Interestingly, loss of Chk2, a DNA damage-activated kinase that arrests the cell cycle and promotes DNA repair and apoptosis, leads to uncontrolled proliferation of intestinal stem cells regardless of their autophagy status. Chk2 accumulates in the nuclei of autophagy-deficient stem cells, raising the possibility that its activation may contribute to the effects of autophagy inhibition in intestinal stem cells. Our study reveals the crucial role of autophagy in preserving proper stem cell function for the continuous renewal of the intestinal epithelium in Drosophila.
Collapse
|
14
|
Yang W, Cho H, Shin HY, Chung JY, Kang ES, Lee EJ, Kim JH. Accumulation of cytoplasmic Cdk1 is associated with cancer growth and survival rate in epithelial ovarian cancer. Oncotarget 2018; 7:49481-49497. [PMID: 27385216 PMCID: PMC5226523 DOI: 10.18632/oncotarget.10373] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Accepted: 06/16/2016] [Indexed: 11/25/2022] Open
Abstract
Cyclin dependent kinase 1 (Cdk1) have previously reported correlation with cancer growth and a key regulator for cell cycle. Mostly, Cdk1′s function of nucleus for cell cycle is well known to be associated with cancer, but cytoplasmic Cdk1′s traits are not clearly identified, yet. We revealed that tissue microarray blocks of epithelial ovarian cancer (n = 249) showed increased level of cytoplasmic Cdk1 (p < 0.001), but not in nucleus (p = 0.192) of histologic cell type independently. On survival analysis, Cdk1 overexpression conferred a significantly worse prognosis in 5-year overall survival (Log-rank p = 0.028, Hazard ratio = 2.016, 95% CI = 1.097 to 4.635). Also, the expression of Cdk1 was increased in ovarian cancer cell lines and Gene Expression Omnibus datasets. When the expression and activity of Cdk1 were inhibited by si-Cdk1 or RO-3306 which is a potent Cdk1 inhibitor, the growth of ovarian cancer was diminished. Moreover, combined treatment with RO-3306 and cisplatin in ovarian cancer significantly elevated anti-cancer effects than single-agent treatment. In conclusion, cytoplasmic Cdk1 expression which was elevated in ovarian cancer predicts a poor overall survival. The inhibition of Cdk1 expression and activity reduced ovarian cancer growth.
Collapse
Affiliation(s)
- Wookyeom Yang
- Department of Obstetrics and Gynecology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea.,Institute of Women's Life Medical Science, Yonsei University College of Medicine, Seoul, Korea
| | - Hanbyoul Cho
- Department of Obstetrics and Gynecology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea.,Institute of Women's Life Medical Science, Yonsei University College of Medicine, Seoul, Korea
| | - Ha-Yeon Shin
- Department of Obstetrics and Gynecology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea.,Institute of Women's Life Medical Science, Yonsei University College of Medicine, Seoul, Korea
| | - Joon-Yong Chung
- Tissue Array Research Program, Laboratory of Pathology, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Eun Suk Kang
- Department of Laboratory Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Eun-Ju Lee
- Department of Obstetrics and Gynecology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea.,Institute of Women's Life Medical Science, Yonsei University College of Medicine, Seoul, Korea
| | - Jae-Hoon Kim
- Department of Obstetrics and Gynecology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea.,Institute of Women's Life Medical Science, Yonsei University College of Medicine, Seoul, Korea
| |
Collapse
|
15
|
Abstract
The activation of the zygotic genome and onset of transcription in blastula embryos is linked to changes in cell behavior and remodeling of the cell cycle and constitutes a transition from exclusive maternal to zygotic control of development. This step in development is referred to as mid-blastula transition and has served as a paradigm for the link between developmental program and cell behavior and morphology. Here, we discuss the mechanism and functional relationships between the zygotic genome activation and cell cycle control during mid-blastula transition with a focus on Drosophila embryos.
Collapse
Affiliation(s)
- Boyang Liu
- Institute for Developmental Biochemistry, Medical School, University of Göttingen, Justus-von-Liebig-Weg11, Göttingen 37077, Germany
| | - Jörg Grosshans
- Institute for Developmental Biochemistry, Medical School, University of Göttingen, Justus-von-Liebig-Weg11, Göttingen 37077, Germany.
| |
Collapse
|
16
|
Al Jord A, Shihavuddin A, Servignat d'Aout R, Faucourt M, Genovesio A, Karaiskou A, Sobczak-Thépot J, Spassky N, Meunier A. Calibrated mitotic oscillator drives motile ciliogenesis. Science 2017; 358:803-806. [PMID: 28982797 DOI: 10.1126/science.aan8311] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Accepted: 09/27/2017] [Indexed: 12/31/2022]
Abstract
Cell division and differentiation depend on massive and rapid organelle remodeling. The mitotic oscillator, centered on the cyclin-dependent kinase 1-anaphase-promoting complex/cyclosome (CDK1-APC/C) axis, spatiotemporally coordinates this reorganization in dividing cells. Here we discovered that nondividing cells could also implement this mitotic clocklike regulatory circuit to orchestrate subcellular reorganization associated with differentiation. We probed centriole amplification in differentiating mouse-brain multiciliated cells. These postmitotic progenitors fine-tuned mitotic oscillator activity to drive the orderly progression of centriole production, maturation, and motile ciliation while avoiding the mitosis commitment threshold. Insufficient CDK1 activity hindered differentiation, whereas excessive activity accelerated differentiation yet drove postmitotic progenitors into mitosis. Thus, postmitotic cells can redeploy and calibrate the mitotic oscillator to uncouple cytoplasmic from nuclear dynamics for organelle remodeling associated with differentiation.
Collapse
Affiliation(s)
- Adel Al Jord
- Institut de Biologie de l'École Normale Supérieure (IBENS), Paris Sciences et Lettres (PSL) Research University, Paris F-75005, France.,CNRS, UMR 8197, Paris F-75005, France.,INSERM, U1024, Paris F-75005, France
| | - Asm Shihavuddin
- Institut de Biologie de l'École Normale Supérieure (IBENS), Paris Sciences et Lettres (PSL) Research University, Paris F-75005, France.,CNRS, UMR 8197, Paris F-75005, France.,INSERM, U1024, Paris F-75005, France
| | - Raphaël Servignat d'Aout
- Institut de Biologie de l'École Normale Supérieure (IBENS), Paris Sciences et Lettres (PSL) Research University, Paris F-75005, France.,CNRS, UMR 8197, Paris F-75005, France.,INSERM, U1024, Paris F-75005, France
| | - Marion Faucourt
- Institut de Biologie de l'École Normale Supérieure (IBENS), Paris Sciences et Lettres (PSL) Research University, Paris F-75005, France.,CNRS, UMR 8197, Paris F-75005, France.,INSERM, U1024, Paris F-75005, France
| | - Auguste Genovesio
- Institut de Biologie de l'École Normale Supérieure (IBENS), Paris Sciences et Lettres (PSL) Research University, Paris F-75005, France.,CNRS, UMR 8197, Paris F-75005, France.,INSERM, U1024, Paris F-75005, France
| | - Anthi Karaiskou
- Sorbonne Universités, Université Pierre et Marie Curie (UPMC) Paris 06, INSERM, Centre de Recherche Saint-Antoine (CRSA), Paris F-75012, France
| | - Joëlle Sobczak-Thépot
- Sorbonne Universités, Université Pierre et Marie Curie (UPMC) Paris 06, INSERM, Centre de Recherche Saint-Antoine (CRSA), Paris F-75012, France
| | - Nathalie Spassky
- Institut de Biologie de l'École Normale Supérieure (IBENS), Paris Sciences et Lettres (PSL) Research University, Paris F-75005, France.,CNRS, UMR 8197, Paris F-75005, France.,INSERM, U1024, Paris F-75005, France
| | - Alice Meunier
- Institut de Biologie de l'École Normale Supérieure (IBENS), Paris Sciences et Lettres (PSL) Research University, Paris F-75005, France. .,CNRS, UMR 8197, Paris F-75005, France.,INSERM, U1024, Paris F-75005, France
| |
Collapse
|
17
|
Deneke VE, Melbinger A, Vergassola M, Di Talia S. Waves of Cdk1 Activity in S Phase Synchronize the Cell Cycle in Drosophila Embryos. Dev Cell 2017; 38:399-412. [PMID: 27554859 DOI: 10.1016/j.devcel.2016.07.023] [Citation(s) in RCA: 94] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Revised: 06/13/2016] [Accepted: 07/26/2016] [Indexed: 01/16/2023]
Abstract
Embryos of most metazoans undergo rapid and synchronous cell cycles following fertilization. While diffusion is too slow for synchronization of mitosis across large spatial scales, waves of Cdk1 activity represent a possible process of synchronization. However, the mechanisms regulating Cdk1 waves during embryonic development remain poorly understood. Using biosensors of Cdk1 and Chk1 activities, we dissect the regulation of Cdk1 waves in the Drosophila syncytial blastoderm. We show that Cdk1 waves are not controlled by the mitotic switch but by a double-negative feedback between Cdk1 and Chk1. Using mathematical modeling and surgical ligations, we demonstrate a fundamental distinction between S phase Cdk1 waves, which propagate as active trigger waves in an excitable medium, and mitotic Cdk1 waves, which propagate as passive phase waves. Our findings show that in Drosophila embryos, Cdk1 positive feedback serves primarily to ensure the rapid onset of mitosis, while wave propagation is regulated by S phase events.
Collapse
Affiliation(s)
- Victoria E Deneke
- Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA
| | - Anna Melbinger
- Department of Physics, University of California San Diego, La Jolla, CA 92093, USA
| | - Massimo Vergassola
- Department of Physics, University of California San Diego, La Jolla, CA 92093, USA
| | - Stefano Di Talia
- Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA.
| |
Collapse
|
18
|
Brose L, Crest J, Tao L, Sullivan W. Polo kinase mediates the phosphorylation and cellular localization of Nuf/FIP3, a Rab11 effector. Mol Biol Cell 2017; 28:1435-1443. [PMID: 28381422 PMCID: PMC5449144 DOI: 10.1091/mbc.e16-04-0236] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Revised: 03/23/2017] [Accepted: 03/31/2017] [Indexed: 01/28/2023] Open
Abstract
Animal cytokinesis involves both actin-myosin-based contraction and vesicle-mediated membrane addition. In many cell types, including early Drosophila embryos, Nuf/FIP3, a Rab11 effector, mediates recycling endosome (RE)-based vesicle delivery to the cytokinesis furrow. Nuf exhibits a cell cycle-regulated concentration at the centrosome that is accompanied by dramatic changes in its phosphorylation state. Here we demonstrate that maximal phosphorylation of Nuf occurs at prophase, when centrosome-associated Nuf disperses throughout the cytoplasm. Accordingly, ectopic Cdk1 activation results in immediate Nuf dispersal from the centrosome. Screening of candidate kinases reveals a specific, dosage-sensitive interaction between Nuf and Polo with respect to Nuf-mediated furrow formation. Inhibiting Polo activity results in Nuf underphosphorylation and prolonged centrosome association. In vitro, Polo directly binds and is required for Nuf phosphorylation at Ser-225 and Thr-227, matching previous in vivo-mapped phosphorylation sites. These results demonstrate a role for Polo kinase in directly mediating Nuf cell cycle-dependent localization.
Collapse
Affiliation(s)
- Lotti Brose
- Department of Molecular, Cell, and Developmental Biology, University of California, Santa Cruz, Santa Cruz, CA 95064
| | - Justin Crest
- Department of Molecular, Cell, and Developmental Biology, University of California, Santa Cruz, Santa Cruz, CA 95064
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720
| | - Li Tao
- Department of Molecular, Cell, and Developmental Biology, University of California, Santa Cruz, Santa Cruz, CA 95064
- Department of Biology, University of Hawaii, Hilo, HI 96720
| | - William Sullivan
- Department of Molecular, Cell, and Developmental Biology, University of California, Santa Cruz, Santa Cruz, CA 95064
| |
Collapse
|
19
|
Ju JF, Hoffmann AA, Zhang YK, Duan XZ, Guo Y, Gong JT, Zhu WC, Hong XY. Wolbachia-induced loss of male fertility is likely related to branch chain amino acid biosynthesis and iLvE in Laodelphax striatellus. INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2017; 85:11-20. [PMID: 28412513 DOI: 10.1016/j.ibmb.2017.04.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Revised: 03/23/2017] [Accepted: 04/10/2017] [Indexed: 06/07/2023]
Abstract
Wolbachia are endosymbionts that infect many species of arthropods and nematodes. Wolbachia-induced cytoplasmic incompatibility (CI) is the most common phenotype in affected hosts, involving embryonic lethality in crosses between Wolbachia-infected males and uninfected females. The molecular mechanisms underlying this phenomenon are currently unclear. Here we examine the molecular correlates of the Wolbachia infection in Laodelphax striatellus (Fallén), an important rice pest, where embryonic lethality is strong and almost complete. We compared the gene expression of 4-day-old Wolbachia-infected and uninfected L. striatellus testes to identify candidate genes for paternal-effect embryonic lethality induction. Based on microarray analysis, iLvE was the most down-regulated gene; this gene mediates branched-chain amino acid (BCAA) biosynthesis and participates in many processes related to reproductive performance. After knocking down iLvE by RNAi in uninfected male L. striatellus, male fertility was reduced, leading to a decrease in embryo hatching rates, but fertility was rescued in crosses between these males and Wolbachia-infected females. Removal of BCAA in chemically-defined diets of uninfected males also led to a loss of male fertility. Low amino acid nutrition may enhance exposure time of sperm to Wolbachia in the testes to affect adult reproduction in L. striatellus by reducing the number of sperm transferred per mating by males. These results indicate that Wolbachia may decrease male fertility in L. striatellus by acting on iLvE, a key factor of BCAA biosynthesis, and delaying sperm maturation.
Collapse
Affiliation(s)
- Jia-Fei Ju
- Department of Entomology, Nanjing Agricultural University, Nanjing, Jiangsu 210095, China.
| | - Ary A Hoffmann
- School of BioSciences, Bio21 Institute, The University of Melbourne, Victoria 3010, Australia.
| | - Yan-Kai Zhang
- Department of Entomology, Nanjing Agricultural University, Nanjing, Jiangsu 210095, China.
| | - Xing-Zhi Duan
- Department of Entomology, Nanjing Agricultural University, Nanjing, Jiangsu 210095, China.
| | - Yan Guo
- Department of Entomology, Nanjing Agricultural University, Nanjing, Jiangsu 210095, China.
| | - Jun-Tao Gong
- Department of Entomology, Nanjing Agricultural University, Nanjing, Jiangsu 210095, China.
| | - Wen-Chao Zhu
- Department of Entomology, Nanjing Agricultural University, Nanjing, Jiangsu 210095, China.
| | - Xiao-Yue Hong
- Department of Entomology, Nanjing Agricultural University, Nanjing, Jiangsu 210095, China.
| |
Collapse
|
20
|
Zhang M, Skirkanich J, Lampson MA, Klein PS. Cell Cycle Remodeling and Zygotic Gene Activation at the Midblastula Transition. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 953:441-487. [DOI: 10.1007/978-3-319-46095-6_9] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
21
|
Deneke VE, Melbinger A, Vergassola M, Di Talia S. Waves of Cdk1 Activity in S Phase Synchronize the Cell Cycle in Drosophila Embryos. Dev Cell 2016. [PMID: 27554859 DOI: 10.1016/j.devcel.2016.07.0240] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/21/2023]
Abstract
Embryos of most metazoans undergo rapid and synchronous cell cycles following fertilization. While diffusion is too slow for synchronization of mitosis across large spatial scales, waves of Cdk1 activity represent a possible process of synchronization. However, the mechanisms regulating Cdk1 waves during embryonic development remain poorly understood. Using biosensors of Cdk1 and Chk1 activities, we dissect the regulation of Cdk1 waves in the Drosophila syncytial blastoderm. We show that Cdk1 waves are not controlled by the mitotic switch but by a double-negative feedback between Cdk1 and Chk1. Using mathematical modeling and surgical ligations, we demonstrate a fundamental distinction between S phase Cdk1 waves, which propagate as active trigger waves in an excitable medium, and mitotic Cdk1 waves, which propagate as passive phase waves. Our findings show that in Drosophila embryos, Cdk1 positive feedback serves primarily to ensure the rapid onset of mitosis, while wave propagation is regulated by S phase events.
Collapse
Affiliation(s)
- Victoria E Deneke
- Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA
| | - Anna Melbinger
- Department of Physics, University of California San Diego, La Jolla, CA 92093, USA
| | - Massimo Vergassola
- Department of Physics, University of California San Diego, La Jolla, CA 92093, USA
| | - Stefano Di Talia
- Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA.
| |
Collapse
|
22
|
Yuan K, Seller CA, Shermoen AW, O'Farrell PH. Timing the Drosophila Mid-Blastula Transition: A Cell Cycle-Centered View. Trends Genet 2016; 32:496-507. [PMID: 27339317 DOI: 10.1016/j.tig.2016.05.006] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2016] [Revised: 05/19/2016] [Accepted: 05/23/2016] [Indexed: 11/18/2022]
Abstract
At the mid-blastula transition (MBT), externally developing embryos refocus from increasing cell number to elaboration of the body plan. Studies in Drosophila reveal a sequence of changes in regulators of Cyclin:Cdk1 that increasingly restricts the activity of this cell cycle kinase to slow cell cycles during early embryogenesis. By reviewing these events, we provide an outline of the mechanisms slowing the cell cycle at and around the time of MBT. The perspectives developed should provide a guiding paradigm for the study of other MBT changes as the embryo transits from maternal control to a regulatory program centered on the expression of zygotic genes.
Collapse
Affiliation(s)
- Kai Yuan
- Department of Biophysics and Biochemistry, University of California San Francisco (UCSF), San Francisco, CA 94158, USA
| | - Charles A Seller
- Department of Biophysics and Biochemistry, University of California San Francisco (UCSF), San Francisco, CA 94158, USA
| | - Antony W Shermoen
- Department of Biophysics and Biochemistry, University of California San Francisco (UCSF), San Francisco, CA 94158, USA
| | - Patrick H O'Farrell
- Department of Biophysics and Biochemistry, University of California San Francisco (UCSF), San Francisco, CA 94158, USA.
| |
Collapse
|
23
|
Microinjection techniques for studying centrosome function in Drosophila melanogaster syncytial embryos. Methods Cell Biol 2015; 129:229-249. [PMID: 26175442 DOI: 10.1016/bs.mcb.2015.03.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
Microinjection is a powerful technique that can be used to study protein function. Early Drosophila embryos are particularly amenable to microinjection due to their large size and their single cell status. Here, we report methods to microinject these embryos with various reagents to study the function of proteins at centrosomes and centrosome function more generally. Although precise details vary between laboratories, many aspects of the process are conserved. We describe the process from setting up a fly cage to imaging the injected embryos on a spinning disk confocal microscope and use specific examples to highlight the potency of this technique.
Collapse
|
24
|
Bergman ZJ, Mclaurin JD, Eritano AS, Johnson BM, Sims AQ, Riggs B. Spatial reorganization of the endoplasmic reticulum during mitosis relies on mitotic kinase cyclin A in the early Drosophila embryo. PLoS One 2015; 10:e0117859. [PMID: 25689737 PMCID: PMC4331435 DOI: 10.1371/journal.pone.0117859] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Accepted: 12/24/2014] [Indexed: 11/19/2022] Open
Abstract
Mitotic cyclin-dependent kinase with their cyclin partners (cyclin:Cdks) are the master regulators of cell cycle progression responsible for regulating a host of activities during mitosis. Nuclear mitotic events, including chromosome condensation and segregation have been directly linked to Cdk activity. However, the regulation and timing of cytoplasmic mitotic events by cyclin:Cdks is poorly understood. In order to examine these mitotic cytoplasmic events, we looked at the dramatic changes in the endoplasmic reticulum (ER) during mitosis in the early Drosophila embryo. The dynamic changes of the ER can be arrested in an interphase state by inhibition of either DNA or protein synthesis. Here we show that this block can be alleviated by micro-injection of Cyclin A (CycA) in which defined mitotic ER clusters gathered at the spindle poles. Conversely, micro-injection of Cyclin B (CycB) did not affect spatial reorganization of the ER, suggesting CycA possesses the ability to initiate mitotic ER events in the cytoplasm. Additionally, RNAi-mediated simultaneous inhibition of all 3 mitotic cyclins (A, B and B3) blocked spatial reorganization of the ER. Our results suggest that mitotic ER reorganization events rely on CycA and that control and timing of nuclear and cytoplasmic events during mitosis may be defined by release of CycA from the nucleus as a consequence of breakdown of the nuclear envelope.
Collapse
Affiliation(s)
- Zane J. Bergman
- Department of Biology, San Francisco State University, 1600 Holloway Ave., San Francisco, California, 94132, United States of America
| | - Justin D. Mclaurin
- Department of Biology, San Francisco State University, 1600 Holloway Ave., San Francisco, California, 94132, United States of America
| | - Anthony S. Eritano
- Department of Biology, San Francisco State University, 1600 Holloway Ave., San Francisco, California, 94132, United States of America
| | - Brittany M. Johnson
- Department of Biology, San Francisco State University, 1600 Holloway Ave., San Francisco, California, 94132, United States of America
| | - Amanda Q. Sims
- Department of Biology, San Francisco State University, 1600 Holloway Ave., San Francisco, California, 94132, United States of America
| | - Blake Riggs
- Department of Biology, San Francisco State University, 1600 Holloway Ave., San Francisco, California, 94132, United States of America
- * E-mail:
| |
Collapse
|
25
|
Yang X, Xu W, Hu Z, Zhang Y, Xu N. Chk1 is required for the metaphase-anaphase transition via regulating the expression and localization of Cdc20 and Mad2. Life Sci 2014; 106:12-8. [PMID: 24747134 DOI: 10.1016/j.lfs.2014.04.011] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2014] [Revised: 03/31/2014] [Accepted: 04/07/2014] [Indexed: 11/16/2022]
Abstract
AIMS The checkpoint kinase 1 (Chk1) functions not only in genotoxic stresses but also in normal cell cycle progression, particularly in the initiation, progression and fidelity of unperturbed mitosis. In this study, we investigated the role of Chk1 in regulating the metaphase-anaphase transition in mammalian cells. MAIN METHODS The mitotic progression was monitored by flow cytometry analysis. The levels of cyclin B1, Cdc20 and Mad2 were measured by Western blotting. Metaphase chromosome alignment and the subcellular localization of Cdc20 and Mad2 were analyzed by immunofluorescence and confocal microscopy. KEY FINDINGS Cyclin B1 degradation and the metaphase-anaphase transition were severely blocked by Chk1 siRNA. Depletion of Chk1 induced chromosome alignment defect in metaphase cells. The kinetochore localization of Cdc20, Mad2 was disrupted in Chk1 depleted cells. Chk1 abrogation also dramatically reduced the protein expression levels of Cdc20 and Mad2. SIGNIFICANCE These results strongly suggest that Chk1 is required for the metaphase-anaphase transition via regulating the subcellular localization and the expression of Cdc20 and Mad2.
Collapse
Affiliation(s)
- Xiaoyun Yang
- School of Life Sciences, Tsinghua University, Beijing 100084, PR China; Key Lab in Healthy Science and Technology, Division of Life Science, Graduate School at Shenzhen, Tsinghua University, Shenzhen, Guangdong Province, PR China
| | - Wei Xu
- School of Life Sciences, Tsinghua University, Beijing 100084, PR China; Key Lab in Healthy Science and Technology, Division of Life Science, Graduate School at Shenzhen, Tsinghua University, Shenzhen, Guangdong Province, PR China
| | - Zuowei Hu
- Department of Clinical Oncology, Wuhan No. 1 Hospital, Wuhan, Hubei Province, PR China
| | - Yaou Zhang
- Key Lab in Healthy Science and Technology, Division of Life Science, Graduate School at Shenzhen, Tsinghua University, Shenzhen, Guangdong Province, PR China
| | - Naihan Xu
- Key Lab in Healthy Science and Technology, Division of Life Science, Graduate School at Shenzhen, Tsinghua University, Shenzhen, Guangdong Province, PR China.
| |
Collapse
|
26
|
Dumollard R, Hebras C, Besnardeau L, McDougall A. Beta-catenin patterns the cell cycle during maternal-to-zygotic transition in urochordate embryos. Dev Biol 2013; 384:331-42. [PMID: 24140189 DOI: 10.1016/j.ydbio.2013.10.007] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2013] [Revised: 09/18/2013] [Accepted: 10/03/2013] [Indexed: 11/18/2022]
Abstract
During the transition from maternal to zygotic control of development, cell cycle length varies in different lineages, and this is important for their fates and functions. The maternal to zygotic transition (MZT) in metazoan embryos involves a profound remodeling of the cell cycle: S phase length increases then G2 is introduced. Although β-catenin is the master regulator of endomesoderm patterning at MZT in all metazoans, the influence of maternal β-catenin on the cell cycle at MZT remains poorly understood. By studying urochordate embryogenesis we found that cell cycle remodeling during MZT begins with the formation of 3 mitotic domains at the 16-cell stage arising from differential S phase lengthening, when endomesoderm is specified. Then, at the 64-cell stage, a G2 phase is introduced in the endoderm lineage during its specification. Strikingly, these two phases of cell cycle remodeling are patterned by β-catenin-dependent transcription. Functional analysis revealed that, at the 16-cell stage, β-catenin speeds up S phase in the endomesoderm. In contrast, two cell cycles later at gastrulation, nuclear β-catenin induces endoderm fate and delays cell division. Such interphase lengthening in invaginating cells is known to be a requisite for gastrulation movements. Therefore, in basal chordates β-catenin has a dual role to specify germ layers and remodel the cell cycle.
Collapse
Affiliation(s)
- Rémi Dumollard
- UMR 7009, UPMC University, Paris 06, France; Centre National de la Recherche (CNRS), Observatoire Océanologique, 06230 Villefranche-sur-Mer, France.
| | | | | | | |
Collapse
|
27
|
Patil M, Pabla N, Dong Z. Checkpoint kinase 1 in DNA damage response and cell cycle regulation. Cell Mol Life Sci 2013; 70:4009-21. [PMID: 23508805 DOI: 10.1007/s00018-013-1307-3] [Citation(s) in RCA: 203] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2012] [Revised: 01/28/2013] [Accepted: 02/18/2013] [Indexed: 12/19/2022]
Abstract
Originally identified as a mediator of DNA damage response (DDR), checkpoint kinase 1 (Chk1) has a broader role in checkpoint activation in DDR and normal cell cycle regulation. Chk1 activation involves phosphorylation at conserved sites. However, recent work has identified a splice variant of Chk1, which may regulate Chk1 in both DDR and normal cell cycle via molecular interaction. Upon activation, Chk1 phosphorylates a variety of substrate proteins, resulting in the activation of DNA damage checkpoints, cell cycle arrest, DNA repair, and/or cell death. Chk1 and its related signaling may be an effective therapeutic target in diseases such as cancer.
Collapse
Affiliation(s)
- Mallikarjun Patil
- Department of Cellular Biology and Anatomy, Georgia Regents University and Charlie Norwood VA Medical Center, 1459 Laney Walker Blvd., Augusta, GA, 30912, USA
| | | | | |
Collapse
|
28
|
Number of nuclear divisions in the Drosophila blastoderm controlled by onset of zygotic transcription. Curr Biol 2013; 23:133-8. [PMID: 23290555 DOI: 10.1016/j.cub.2012.12.013] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2012] [Revised: 11/25/2012] [Accepted: 12/10/2012] [Indexed: 01/10/2023]
Abstract
The cell number of the early Drosophila embryo is determined by exactly 13 rounds of synchronous nuclear divisions, allowing cellularization and formation of the embryonic epithelium. The pause in G2 in cycle 14 is controlled by multiple pathways, such as activation of DNA repair checkpoint, progression through S phase, and inhibitory phosphorylation of Cdk1, involving the genes grapes, mei41, and wee1. In addition, degradation of maternal RNAs and zygotic gene expression are involved. The zinc finger Vielfältig (Vfl) controls expression of many early zygotic genes, including the mitotic inhibitor frühstart. The functional relationship of these pathways and the mechanism for triggering the cell-cycle pause have remained unclear. Here, we show that a novel single-nucleotide mutation in the 3' UTR of the RNPII215 gene leads to a reduced number of nuclear divisions that is accompanied by premature transcription of early zygotic genes and cellularization. The reduced number of nuclear divisions in mutant embryos depends on the transcription factor Vfl and on zygotic gene expression, but not on grapes, the mitotic inhibitor Frühstart, and the nucleocytoplasmic ratio. We propose that activation of zygotic gene expression is the trigger that determines the timely and concerted cell-cycle pause and cellularization.
Collapse
|
29
|
Farrell JA, O'Farrell PH. Mechanism and regulation of Cdc25/Twine protein destruction in embryonic cell-cycle remodeling. Curr Biol 2013; 23:118-26. [PMID: 23290551 DOI: 10.1016/j.cub.2012.11.036] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2012] [Revised: 10/23/2012] [Accepted: 11/19/2012] [Indexed: 10/27/2022]
Abstract
BACKGROUND In Drosophila embryos, the midblastula transition (MBT) dramatically remodels the cell cycle during the 14(th) interphase. Before the MBT, each cycle is composed of only a short S phase and mitosis. At the MBT, S phase is dramatically lengthened by the onset of late replication, and a G2 phase is introduced. Both changes set the stage for gastrulation and require downregulation of Cdc25 phosphatase, which was previously attributed to the elimination of its transcripts at the MBT. RESULTS Premature removal of cdc25 transcripts by RNAi did not affect progression to the MBT. Instead, an antibody against the Cdc25 isoform Twine showed that Twine protein was abundant and stable until the MBT, when it was destabilized and rapidly eliminated. Persistence of pre-MBT levels of Twine was sufficient to prevent cell-cycle slowing. Twine protein destruction was timed by the nucleocytoplasmic ratio and depended on the activation of zygotic transcription at the MBT, including expression of the gene tribbles, whose activity was sufficient to trigger Twine destruction and was required for prompt Twine disappearance. CONCLUSIONS We propose that the developmentally regulated destruction of Twine protein is a critical switch that contributes to the cell-cycle change at the MBT, including the addition of a G2 phase and onset of late replication. Moreover, we show that this destruction is triggered by the nucleocytoplasmic ratio-dependent onset of zygotic transcription of tribbles and other unknown genes.
Collapse
Affiliation(s)
- Jeffrey A Farrell
- Department of Biochemistry, University of California, San Francisco, San Francisco, CA 94158, USA
| | | |
Collapse
|
30
|
Yuan K, Farrell JA, O'Farrell PH. Different cyclin types collaborate to reverse the S-phase checkpoint and permit prompt mitosis. ACTA ACUST UNITED AC 2012; 198:973-80. [PMID: 22965907 PMCID: PMC3444785 DOI: 10.1083/jcb.201205007] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Different cyclin types have distinct abilities to reverse the S-phase checkpoint, and timely entry into mitosis after embryonic S phase requires collaborative action of multiple cyclin types. Precise timing coordinates cell proliferation with embryonic morphogenesis. As Drosophila melanogaster embryos approach cell cycle 14 and the midblastula transition, rapid embryonic cell cycles slow because S phase lengthens, which delays mitosis via the S-phase checkpoint. We probed the contributions of each of the three mitotic cyclins to this timing of interphase duration. Each pairwise RNA interference knockdown of two cyclins lengthened interphase 13 by introducing a G2 phase of a distinct duration. In contrast, pairwise cyclin knockdowns failed to introduce a G2 in embryos that lacked an S-phase checkpoint. Thus, the single remaining cyclin is sufficient to induce early mitotic entry, but reversal of the S-phase checkpoint is compromised by pairwise cyclin knockdown. Manipulating cyclin levels revealed that the diversity of cyclin types rather than cyclin level influenced checkpoint reversal. We conclude that different cyclin types have distinct abilities to reverse the checkpoint but that they collaborate to do so rapidly.
Collapse
Affiliation(s)
- Kai Yuan
- Department of Biochemistry, University of California, San Francisco, San Francisco, CA 94158, USA
| | | | | |
Collapse
|
31
|
Farrell JA, Shermoen AW, Yuan K, O'Farrell PH. Embryonic onset of late replication requires Cdc25 down-regulation. Genes Dev 2012; 26:714-25. [PMID: 22431511 DOI: 10.1101/gad.186429.111] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The Drosophila midblastula transition (MBT), a major event in embryogenesis, remodels and slows the cell cycle. In the pre-MBT cycles, all genomic regions replicate simultaneously in rapid S phases that alternate with mitosis, skipping gap phases. At the MBT, down-regulation of Cdc25 phosphatase and the resulting inhibitory phosphorylation of the mitotic kinase Cdk1 create a G2 pause in interphase 14. However, an earlier change in interphase 14 is the prolongation of S phase. While the signals modifying S phase are unknown, the onset of late replication-where replication of constitutively heterochromatic satellite sequences is delayed-extends S-phase 14. We injected Cdc25 mRNA to bypass the developmentally programmed down-regulation of Cdc25 at the MBT. Introduction of either Cdc25 isoform (String or Twine) or enhanced Cdk1 activity triggered premature replication of late-replicating sequences, even after their specification, and thereby shortened S phase. Reciprocally, reduction of Cdk1 activity by knockdown of mitotic cyclins extended pre-MBT S phase. These findings suggest that high Cdc25 and Cdk1 contribute to the speed of the rapid, pre-MBT S phases and that down-regulation of these activities plays a broader role in MBT-associated changes than was previously suspected.
Collapse
Affiliation(s)
- Jeffrey A Farrell
- Department of Biochemistry, University of California at San Francisco, San Francisco, California 94143, USA
| | | | | | | |
Collapse
|
32
|
Fasulo B, Koyama C, Yu KR, Homola EM, Hsieh TS, Campbell SD, Sullivan W. Chk1 and Wee1 kinases coordinate DNA replication, chromosome condensation, and anaphase entry. Mol Biol Cell 2012; 23:1047-57. [PMID: 22262459 PMCID: PMC3302732 DOI: 10.1091/mbc.e11-10-0832] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2022] Open
Abstract
New chromosome condensation checkpoints are identified. S-phase and topoisomerase inhibitors delay chromosome condensation. These delays require chk1 and wee1. Inhibitors causing defects in chromosome condensation/congression on the metaphase plate delay anaphase entry. wee1 and not the spindle assembly checkpoint mediates the delay. Defects in DNA replication and chromosome condensation are common phenotypes in cancer cells. A link between replication and condensation has been established, but little is known about the role of checkpoints in monitoring chromosome condensation. We investigate this function by live analysis, using the rapid division cycles in the early Drosophila embryo. We find that S-phase and topoisomerase inhibitors delay both the initiation and the rate of chromosome condensation. These cell cycle delays are mediated by the cell cycle kinases chk1 and wee1. Inhibitors that cause severe defects in chromosome condensation and congression on the metaphase plate result in delayed anaphase entry. These delays are mediated by wee1 and are not the result of spindle assembly checkpoint activation. In addition, we provide the first detailed live analysis of the direct effect of widely used anticancer agents (aclarubicin, ICRF-193, VM26, doxorubicin, camptothecin, aphidicolin, hydroxyurea, cisplatin, mechlorethamine and x-rays) on key nuclear and cytoplasmic cell cycle events.
Collapse
Affiliation(s)
- Barbara Fasulo
- Sinsheimer Laboratories, Department of Molecular, Cellular and Developmental Biology, University of California, Santa Cruz, Santa Cruz, CA 95064, USA.
| | | | | | | | | | | | | |
Collapse
|
33
|
Molecular and genetic analysis of the Drosophila model of fragile X syndrome. Results Probl Cell Differ 2012; 54:119-56. [PMID: 22009350 DOI: 10.1007/978-3-642-21649-7_7] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
The Drosophila genome contains most genes known to be involved in heritable disease. The extraordinary genetic malleability of Drosophila, coupled to sophisticated imaging, electrophysiology, and behavioral paradigms, has paved the way for insightful mechanistic studies on the causes of developmental and neurological disease as well as many possible interventions. Here, we focus on one of the most advanced examples of Drosophila genetic disease modeling, the Drosophila model of Fragile X Syndrome, which for the past decade has provided key advances into the molecular, cellular, and behavioral defects underlying this devastating disorder. We discuss the multitude of RNAs and proteins that interact with the disease-causing FMR1 gene product, whose function is conserved from Drosophila to human. In turn, we consider FMR1 mechanistic relationships in non-neuronal tissues (germ cells and embryos), peripheral motor and sensory circuits, and central brain circuits involved in circadian clock activity and learning/memory.
Collapse
|
34
|
Amcheslavsky A, Ito N, Jiang J, Ip YT. Tuberous sclerosis complex and Myc coordinate the growth and division of Drosophila intestinal stem cells. ACTA ACUST UNITED AC 2011; 193:695-710. [PMID: 21555458 PMCID: PMC3166862 DOI: 10.1083/jcb.201103018] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Excessive cell growth in Drosophila intestinal stem cells lacking TSC blocks further cell division. Intestinal stem cells (ISCs) in the adult Drosophila melanogaster midgut can respond to damage and support repair. We demonstrate in this paper that the tuberous sclerosis complex (TSC) plays a critical role in balancing ISC growth and division. Previous studies have shown that imaginal disc cells that are mutant for TSC have increased rates of growth and division. However, we report in this paper that loss of TSC in the adult Drosophila midgut results in the formation of much larger ISCs that have halted cell division. These mutant ISCs expressed proper stem cell markers, did not differentiate, and had defects in multiple steps of the cell cycle. Slowing the growth by feeding rapamycin or reducing Myc was sufficient to rescue the division defect. The TSC mutant guts had a thinner epithelial structure than wild-type tissues, and the mutant flies were more susceptible to tissue damage. Therefore, we have uncovered a context-dependent phenotype of TSC mutants in adult ISCs, such that the excessive growth leads to inhibition of division.
Collapse
Affiliation(s)
- Alla Amcheslavsky
- University of Massachusetts Medical School, Worcester, MA 01605, USA
| | | | | | | |
Collapse
|
35
|
Zheng Y, Ren PP, Wang JL, Wang YF. Wolbachia-induced cytoplasmic incompatibility is associated with decreased Hira expression in male Drosophila. PLoS One 2011; 6:e19512. [PMID: 21559343 PMCID: PMC3084885 DOI: 10.1371/journal.pone.0019512] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2010] [Accepted: 03/31/2011] [Indexed: 11/19/2022] Open
Abstract
Background Wolbachia are obligate endosymbiotic bacteria that infect numerous species of arthropods and nematodes. Wolbachia can induce several reproductive phenotypes in their insect hosts including feminization, male-killing, parthenogenesis and cytoplasmic incompatibility (CI). CI is the most common phenotype and occurs when Wolbachia-infected males mate with uninfected females resulting in no or very low numbers of viable offspring. However, matings between males and females infected with the same strain of Wolbachia result in viable progeny. Despite substantial scientific effort, the molecular mechanisms underlying CI are currently unknown. Methodology/Principal Findings Gene expression studies were undertaken in Drosophila melanogaster and D. simulans which display differential levels of CI using quantitative RT-PCR. We show that Hira expression is correlated with the induction of CI and occurs in a sex-specific manner. Hira expression is significantly lower in males which induce strong CI when compared to males inducing no CI or Wolbachia-uninfected males. A reduction in Hira expression is also observed in 1-day-old males that induce stronger CI compared to 5-day-old males that induce weak or no CI. In addition, Hira mutated D. melanogaster males mated to uninfected females result in significantly decreased hatch rates comparing with uninfected crosses. Interestingly, wMel-infected females may rescue the hatch rates. An obvious CI phenotype with chromatin bridges are observed in the early embryo resulting from Hira mutant fertilization, which strongly mimics the defects associated with CI. Conclusions/Significance Our results suggest Wolbachia-induced CI in Drosophila occurs due to a reduction in Hira expression in Wolbachia-infected males leading to detrimental effects on sperm fertility resulting in embryo lethality. These results may help determine the underlying mechanism of CI and provide further insight in to the important role Hira plays in the interaction of Wolbachia and its insect host.
Collapse
Affiliation(s)
- Ya Zheng
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, College of Life Science, Huazhong Normal University, Wuhan, People's Republic of China
| | - Pan-Pan Ren
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, College of Life Science, Huazhong Normal University, Wuhan, People's Republic of China
| | - Jia-Lin Wang
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, College of Life Science, Huazhong Normal University, Wuhan, People's Republic of China
| | - Yu-Feng Wang
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, College of Life Science, Huazhong Normal University, Wuhan, People's Republic of China
- * E-mail:
| |
Collapse
|
36
|
Holt JE, Tran SMT, Stewart JL, Minahan K, García-Higuera I, Moreno S, Jones KT. The APC/C activator FZR1 coordinates the timing of meiotic resumption during prophase I arrest in mammalian oocytes. Development 2011; 138:905-13. [DOI: 10.1242/dev.059022] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
FZR1, an activator of the anaphase-promoting complex/cyclosome (APC/C), is recognized for its roles in the mitotic cell cycle. To examine its meiotic function in females we generated an oocyte-specific knockout of the Fzr1 gene (Fzr1Δ/Δ). The total number of fully grown oocytes enclosed in cumulus complexes was 35-40% lower in oocytes from Fzr1Δ/Δ mice and there was a commensurate rise in denuded, meiotically advanced and/or fragmented oocytes. The ability of Fzr1Δ/Δ oocytes to remain prophase I/germinal vesicle (GV) arrested in vitro was also compromised, despite the addition of the phosphodiesterase milrinone. Meiotic competency of smaller diameter oocytes was also accelerated by Fzr1 loss. Cyclin B1 levels were elevated ~5-fold in Fzr1Δ/Δ oocytes, whereas securin and CDC25B, two other APC/CFZR1 substrates, were unchanged. Cyclin B1 overexpression can mimic the effects of Fzr1 loss on GV arrest and here we show that cyclin B1 knockdown in Fzr1Δ/Δ oocytes affects the timing of meiotic resumption. Therefore, the effects of Fzr1 loss are mediated, at least in part, by raised cyclin B1. Thus, APC/CFZR1 activity is required to repress cyclin B1 levels in oocytes during prophase I arrest in the ovary, thereby maintaining meiotic quiescence until hormonal cues trigger resumption.
Collapse
Affiliation(s)
- Janet E. Holt
- School of Biomedical Sciences, University of Newcastle, Callaghan, NSW 2308, Australia
| | - Suzanne M.-T. Tran
- School of Biomedical Sciences, University of Newcastle, Callaghan, NSW 2308, Australia
| | - Jessica L. Stewart
- School of Biomedical Sciences, University of Newcastle, Callaghan, NSW 2308, Australia
| | - Kyra Minahan
- School of Biomedical Sciences, University of Newcastle, Callaghan, NSW 2308, Australia
| | - Irene García-Higuera
- Instituto de Biología Molecular y Celular del Cáncer, CSIC/Salamanca University, Campus Miguel de Unamuno, 37007 Salamanca, Spain
| | - Sergio Moreno
- Instituto de Biología Molecular y Celular del Cáncer, CSIC/Salamanca University, Campus Miguel de Unamuno, 37007 Salamanca, Spain
| | - Keith T. Jones
- School of Biomedical Sciences, University of Newcastle, Callaghan, NSW 2308, Australia
| |
Collapse
|
37
|
Papoulas O, Monzo KF, Cantin GT, Ruse C, Yates JR, Ryu YH, Sisson JC. dFMRP and Caprin, translational regulators of synaptic plasticity, control the cell cycle at the Drosophila mid-blastula transition. Development 2010; 137:4201-9. [PMID: 21068064 DOI: 10.1242/dev.055046] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
The molecular mechanisms driving the conserved metazoan developmental shift referred to as the mid-blastula transition (MBT) remain mysterious. Typically, cleavage divisions give way to longer asynchronous cell cycles with the acquisition of a gap phase. In Drosophila, rapid synchronous nuclear divisions must pause at the MBT to allow the formation of a cellular blastoderm through a special form of cytokinesis termed cellularization. Drosophila Fragile X mental retardation protein (dFMRP; FMR1), a transcript-specific translational regulator, is required for cellularization. The role of FMRP has been most extensively studied in the nervous system because the loss of FMRP activity in neurons causes the misexpression of specific mRNAs required for synaptic plasticity, resulting in mental retardation and autism in humans. Here, we show that in the early embryo dFMRP associates specifically with Caprin, another transcript-specific translational regulator implicated in synaptic plasticity, and with eIF4G, a key regulator of translational initiation. dFMRP and Caprin collaborate to control the cell cycle at the MBT by directly mediating the normal repression of maternal Cyclin B mRNA and the activation of zygotic frühstart mRNA. These findings identify two new targets of dFMRP regulation and implicate conserved translational regulatory mechanisms in processes as diverse as learning, memory and early embryonic development.
Collapse
Affiliation(s)
- Ophelia Papoulas
- The Section of MCD Biology and Institute for Cellular and Molecular Biology, The University of Texas at Austin, TX 78712, USA.
| | | | | | | | | | | | | |
Collapse
|
38
|
Malzer E, Daly ML, Moloney A, Sendall TJ, Thomas SE, Ryder E, Ryoo HD, Crowther DC, Lomas DA, Marciniak SJ. Impaired tissue growth is mediated by checkpoint kinase 1 (CHK1) in the integrated stress response. J Cell Sci 2010; 123:2892-900. [PMID: 20682638 DOI: 10.1242/jcs.070078] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The integrated stress response (ISR) protects cells from numerous forms of stress and is involved in the growth of solid tumours; however, it is unclear how the ISR acts on cellular proliferation. We have developed a model of ISR signalling with which to study its effects on tissue growth. Overexpression of the ISR kinase PERK resulted in a striking atrophic eye phenotype in Drosophila melanogaster that could be rescued by co-expressing the eIF2alpha phosphatase GADD34. A genetic screen of 3000 transposon insertions identified grapes, the gene that encodes the Drosophila orthologue of checkpoint kinase 1 (CHK1). Knockdown of grapes by RNAi rescued eye development despite ongoing PERK activation. In mammalian cells, CHK1 was activated by agents that induce ER stress, which resulted in a G2 cell cycle delay. PERK was both necessary and sufficient for CHK1 activation. These findings indicate that non-genotoxic misfolded protein stress accesses DNA-damage-induced cell cycle checkpoints to couple the ISR to cell cycle arrest.
Collapse
Affiliation(s)
- Elke Malzer
- Department of Medicine, University of Cambridge, Cambridge Institute for Medical Research (CIMR), Wellcome Trust/MRC Building, Hills Road, Cambridge, CB2 0XY, UK
| | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Gavet O, Pines J. Activation of cyclin B1-Cdk1 synchronizes events in the nucleus and the cytoplasm at mitosis. ACTA ACUST UNITED AC 2010; 189:247-59. [PMID: 20404109 PMCID: PMC2856909 DOI: 10.1083/jcb.200909144] [Citation(s) in RCA: 219] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
The cyclin B-Cdk1 kinase triggers mitosis in most eukaryotes. In animal cells, cyclin B shuttles between the nucleus and cytoplasm in interphase before rapidly accumulating in the nucleus at prophase, which promotes disassembly of the nuclear lamina and nuclear envelope breakdown (NEBD). What triggers the nuclear accumulation of cyclin B1 is presently unclear, although the prevailing view is that the Plk1 kinase inhibits its nuclear export. In this study, we use a biosensor specific for cyclin B1-Cdk1 activity to show that activating cyclin B1-Cdk1 immediately triggers its rapid accumulation in the nucleus through a 40-fold increase in nuclear import that remains dependent on Cdk1 activity until NEBD. Nevertheless, a substantial proportion of cyclin B1-Cdk1 remains in the cytoplasm. The increase in nuclear import is driven by changes in the nuclear import machinery that require neither Plk1 nor inhibition of nuclear export. Thus, the intrinsic link between cyclin B1-Cdk1 activation and its rapid nuclear import inherently coordinates the reorganization of the nucleus and the cytoplasm at mitotic entry.
Collapse
Affiliation(s)
- Olivier Gavet
- The Wellcome Trust/Cancer Research UK Gurdon Institute, University of Cambridge, Cambridge CB2 1QN, England, UK.
| | | |
Collapse
|
40
|
McCleland ML, Farrell JA, O'Farrell PH. Influence of cyclin type and dose on mitotic entry and progression in the early Drosophila embryo. ACTA ACUST UNITED AC 2009; 184:639-46. [PMID: 19273612 PMCID: PMC2686416 DOI: 10.1083/jcb.200810012] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Cyclins are key cell cycle regulators, yet few analyses test their role in timing the events that they regulate. We used RNA interference and real-time visualization in embryos to define the events regulated by each of the three mitotic cyclins of Drosophila melanogaster, CycA, CycB, and CycB3. Each individual and pairwise knockdown results in distinct mitotic phenotypes. For example, mitosis without metaphase occurs upon knockdown of CycA and CycB. To separate the role of cyclin levels from the influences of cyclin type, we knocked down two cyclins and reduced the gene dose of the one remaining cyclin. This reduction did not prolong interphase but instead interrupted mitotic progression. Mitotic prophase chromosomes formed, centrosomes divided, and nuclei exited mitosis without executing later events. This prompt but curtailed mitosis shows that accumulation of cyclin function does not directly time mitotic entry in these early embryonic cycles and that cyclin function can be sufficient for some mitotic events although inadequate for others.
Collapse
Affiliation(s)
- Mark L McCleland
- Department of Biochemistry and Biophysics, University of California, San Francisco, CA 94143, USA
| | | | | |
Collapse
|
41
|
Wolbachia-mediated cytoplasmic incompatibility is associated with impaired histone deposition in the male pronucleus. PLoS Pathog 2009; 5:e1000343. [PMID: 19300496 PMCID: PMC2652114 DOI: 10.1371/journal.ppat.1000343] [Citation(s) in RCA: 118] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2008] [Accepted: 02/20/2009] [Indexed: 01/24/2023] Open
Abstract
Wolbachia is a bacteria endosymbiont that rapidly infects insect populations through a mechanism known as cytoplasmic incompatibility (CI). In CI, crosses between Wolbachia-infected males and uninfected females produce severe cell cycle defects in the male pronucleus resulting in early embryonic lethality. In contrast, viable progeny are produced when both parents are infected (the Rescue cross). An important consequence of CI–Rescue is that infected females have a selective advantage over uninfected females facilitating the rapid spread of Wolbachia through insect populations. CI disrupts a number of prophase and metaphase events in the male pronucleus, including Cdk1 activation, chromosome condensation, and segregation. Here, we demonstrate that CI disrupts earlier interphase cell cycle events. Specifically, CI delays the H3.3 and H4 deposition that occurs immediately after protamine removal from the male pronucleus. In addition, we find prolonged retention of the replication factor PCNA in the male pronucleus into metaphase, indicating progression into mitosis with incompletely replicated DNA. We propose that these CI-induced interphase defects in de novo nucleosome assembly and replication are the cause of the observed mitotic condensation and segregation defects. In addition, these interphase chromosome defects likely activate S-phase checkpoints, accounting for the previously described delays in Cdk1 activation. These results have important implications for the mechanism of Rescue and other Wolbachia-induced phenotypes. Wolbachia are among the most successful of all intracellular bacteria, infecting an estimated 65% of insect species. Wolbachia are also present in filarial nematodes and are the cause of African river blindness. Wolbachia's success is due in part to its ability to induce a conditional form of sterility known as cytoplasmic incompatibility (CI), endowing infected females with a tremendous selective advantage. CI results in the severe reduction in progeny from crosses between uninfected females and Wolbachia-infected males. However, Wolbachia-infected females can mate with either infected or uninfected males with no reduction in progeny. CI may drive speciation and is intensively being pursued as a means to control insect-borne human disease. In spite of its biological and medical significance, the molecular basis of CI is not understood. We take advantage of newly generated chromatin reagents to demonstrate that prior to the well-documented defects in chromosome condensation and segregation, CI produces a delay in recruiting the replication-independent histone H3.3/H4 complex to the male pronucleus. There is great interest in histone H3.3 because of its general role in transcription and in remodeling of the sperm chromatin following fertilization. In addition, these findings may provide insight into other Wolbachia–host interactions such as CI–Rescue and male-killing.
Collapse
|