1
|
Connors CQ, Mauro MS, Wiles JT, Countryman AD, Martin SL, Lacroix B, Shirasu-Hiza M, Dumont J, Kasza KE, Davies TR, Canman JC. Germ fate determinants protect germ precursor cell division by reducing septin and anillin levels at the cell division plane. Mol Biol Cell 2024; 35:ar94. [PMID: 38696255 PMCID: PMC11244169 DOI: 10.1091/mbc.e24-02-0096-t] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 04/22/2024] [Accepted: 04/26/2024] [Indexed: 05/04/2024] Open
Abstract
Animal cell cytokinesis, or the physical division of one cell into two, is thought to be driven by constriction of an actomyosin contractile ring at the division plane. The mechanisms underlying cell type-specific differences in cytokinesis remain unknown. Germ cells are totipotent cells that pass genetic information to the next generation. Previously, using formincyk-1(ts) mutant Caenorhabditis elegans 4-cell embryos, we found that the P2 germ precursor cell is protected from cytokinesis failure and can divide with greatly reduced F-actin levels at the cell division plane. Here, we identified two canonical germ fate determinants required for P2-specific cytokinetic protection: PIE-1 and POS-1. Neither has been implicated previously in cytokinesis. These germ fate determinants protect P2 cytokinesis by reducing the accumulation of septinUNC-59 and anillinANI-1 at the division plane, which here act as negative regulators of cytokinesis. These findings may provide insight into the regulation of cytokinesis in other cell types, especially in stem cells with high potency.
Collapse
Affiliation(s)
- Caroline Q. Connors
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY 10032
| | - Michael S. Mauro
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY 10032
| | - J. Tristian Wiles
- Department of Biological Sciences, Columbia University, New York, NY 10027
| | | | - Sophia L. Martin
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY 10032
| | - Benjamin Lacroix
- Université Paris Cité, CNRS, Institut Jacques Monod, F-75013 Paris, France
- Université de Montpellier, CNRS, Centre de Recherche en Biologie Cellulaire de Montpellier, UMR 5237 Montpellier, France
| | - Mimi Shirasu-Hiza
- Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY 10032
| | - Julien Dumont
- Université Paris Cité, CNRS, Institut Jacques Monod, F-75013 Paris, France
| | - Karen E. Kasza
- Department of Mechanical Engineering, Columbia University, New York, NY 10027
| | - Timothy R. Davies
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY 10032
- Department of Biosciences, Durham University, Durham DH1 3LE, UK
| | - Julie C. Canman
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY 10032
| |
Collapse
|
2
|
Connors CQ, Mauro MS, Tristian Wiles J, Countryman AD, Martin SL, Lacroix B, Shirasu-Hiza M, Dumont J, Kasza KE, Davies TR, Canman JC. Germ fate determinants protect germ precursor cell division by restricting septin and anillin levels at the division plane. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.17.566773. [PMID: 38014027 PMCID: PMC10680835 DOI: 10.1101/2023.11.17.566773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
Animal cell cytokinesis, or the physical division of one cell into two, is thought to be driven by constriction of an actomyosin contractile ring at the division plane. The mechanisms underlying cell type-specific differences in cytokinesis remain unknown. Germ cells are totipotent cells that pass genetic information to the next generation. Previously, using formin cyk-1 (ts) mutant C. elegans embryos, we found that the P2 germ precursor cell is protected from cytokinesis failure and can divide without detectable F-actin at the division plane. Here, we identified two canonical germ fate determinants required for P2-specific cytokinetic protection: PIE-1 and POS-1. Neither has been implicated previously in cytokinesis. These germ fate determinants protect P2 cytokinesis by reducing the accumulation of septin UNC-59 and anillin ANI-1 at the division plane, which here act as negative regulators of cytokinesis. These findings may provide insight into cytokinetic regulation in other cell types, especially in stem cells with high potency.
Collapse
|
3
|
Iegiani G, Ferraro A, Pallavicini G, Di Cunto F. The impact of TP53 activation and apoptosis in primary hereditary microcephaly. Front Neurosci 2023; 17:1220010. [PMID: 37457016 PMCID: PMC10338886 DOI: 10.3389/fnins.2023.1220010] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 06/12/2023] [Indexed: 07/18/2023] Open
Abstract
Autosomal recessive primary microcephaly (MCPH) is a constellation of disorders that share significant brain size reduction and mild to moderate intellectual disability, which may be accompanied by a large variety of more invalidating clinical signs. Extensive neural progenitor cells (NPC) proliferation and differentiation are essential to determine brain final size. Accordingly, the 30 MCPH loci mapped so far (MCPH1-MCPH30) encode for proteins involved in microtubule and spindle organization, centriole biogenesis, nuclear envelope, DNA replication and repair, underscoring that a wide variety of cellular processes is required for sustaining NPC expansion during development. Current models propose that altered balance between symmetric and asymmetric division, as well as premature differentiation, are the main mechanisms leading to MCPH. Although studies of cellular alterations in microcephaly models have constantly shown the co-existence of high DNA damage and apoptosis levels, these mechanisms are less considered as primary factors. In this review we highlight how the molecular and cellular events produced by mutation of the majority of MCPH genes may converge on apoptotic death of NPCs and neurons, via TP53 activation. We propose that these mechanisms should be more carefully considered in the alterations of the sophisticated equilibrium between proliferation, differentiation and death produced by MCPH gene mutations. In consideration of the potential druggability of cell apoptotic pathways, a better understanding of their role in MCPH may significantly facilitate the development of translational approaches.
Collapse
Affiliation(s)
- Giorgia Iegiani
- Department of Neuroscience ‘Rita Levi Montalcini’, University of Turin, Turin, Italy
- Neuroscience Institute Cavalieri Ottolenghi, Turin, Italy
| | - Alessia Ferraro
- Department of Neuroscience ‘Rita Levi Montalcini’, University of Turin, Turin, Italy
- Neuroscience Institute Cavalieri Ottolenghi, Turin, Italy
| | - Gianmarco Pallavicini
- Department of Neuroscience ‘Rita Levi Montalcini’, University of Turin, Turin, Italy
- Neuroscience Institute Cavalieri Ottolenghi, Turin, Italy
| | - Ferdinando Di Cunto
- Department of Neuroscience ‘Rita Levi Montalcini’, University of Turin, Turin, Italy
- Neuroscience Institute Cavalieri Ottolenghi, Turin, Italy
| |
Collapse
|
4
|
Li X, Heng BC, Bai Y, Wang Q, Gao M, He Y, Zhang X, Deng X, Zhang X. Electrical charge on ferroelectric nanocomposite membranes enhances SHED neural differentiation. Bioact Mater 2023; 20:81-92. [PMID: 35633875 PMCID: PMC9131252 DOI: 10.1016/j.bioactmat.2022.05.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 05/04/2022] [Accepted: 05/05/2022] [Indexed: 11/16/2022] Open
Abstract
Stem cells from human exfoliated deciduous teeth (SHED) uniquely exhibit high proliferative and neurogenic potential. Charged biomaterials have been demonstrated to promote neural differentiation of stem cells, but the dose-response effect of electrical stimuli from these materials on neural differentiation of SHED remains to be elucidated. Here, by utilizing different annealing temperatures prior to corona poling treatment, BaTiO3/P(VDF-TrFE) ferroelectric nanocomposite membranes with varying charge polarization intensity (d33 ≈ 0, 4, 12 and 19 pC N−1) were fabricated. Enhanced expression of neural markers, increased cell elongation and more prominent neurite outgrowths were observed with increasing surface charge of the nanocomposite membrane indicating a dose-response effect of surface electrical charge on SHED neural differentiation. Further investigations of the underlying molecular mechanisms revealed that intracellular calcium influx, focal adhesion formation, FAK-ERK mechanosensing pathway and neurogenic-related ErbB signaling pathway were implicated in the enhancement of SHED neural differentiation by surface electrical charge. Hence, this study confirms the dose-response effect of biomaterial surface charge on SHED neural differentiation and provides preliminary insights into the molecular mechanisms and signaling pathways involved. Membrane surface charge can be precisely controlled by adjusting annealing temperature and corona poling parameters. Both earlier and later neurogenic differentiation of SHED appear to be dose-dependently enhanced by surface charge. Underlying molecular mechanisms may involve intracellular Ca2+ influx, focal adhesion formation, FAK-ERK and ErbB signaling.
Collapse
Affiliation(s)
- Xiaochan Li
- Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, 100081, PR China
- Department of Dental Materials & Dental Medical Devices Testing Center, Peking University School and Hospital of Stomatology, Beijing, 100081, PR China
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, Beijing, 100081, PR China
| | - Boon Chin Heng
- Central Laboratory, Peking University School and Hospital of Stomatology, Beijing, 100081, PR China
| | - Yunyang Bai
- Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, 100081, PR China
| | - Qianqian Wang
- Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, 100081, PR China
| | - Min Gao
- Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, 100081, PR China
| | - Ying He
- Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, 100081, PR China
| | - Xinwen Zhang
- Center of Implant Dentistry, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang, 110002, PR China
- Corresponding author.
| | - Xuliang Deng
- Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, 100081, PR China
- National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, NMPA Key Laboratory for Dental Materials, Beijing Laboratory of Biomedical Materials, Peking University School and Hospital of Stomatology, Beijing, 100081, PR China
- Corresponding author. Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, 100081, PR China.
| | - Xuehui Zhang
- Department of Dental Materials & Dental Medical Devices Testing Center, Peking University School and Hospital of Stomatology, Beijing, 100081, PR China
- National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, NMPA Key Laboratory for Dental Materials, Beijing Laboratory of Biomedical Materials, Peking University School and Hospital of Stomatology, Beijing, 100081, PR China
- Corresponding author. Department of Dental Materials & Dental Medical Devices Testing Center, Peking University School and Hospital of Stomatology, Beijing, 100081, PR China.
| |
Collapse
|
5
|
Boda E, Lorenzati M, Parolisi R, Harding B, Pallavicini G, Bonfanti L, Moccia A, Bielas S, Di Cunto F, Buffo A. Molecular and functional heterogeneity in dorsal and ventral oligodendrocyte progenitor cells of the mouse forebrain in response to DNA damage. Nat Commun 2022; 13:2331. [PMID: 35484145 PMCID: PMC9051058 DOI: 10.1038/s41467-022-30010-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Accepted: 04/07/2022] [Indexed: 11/09/2022] Open
Abstract
In the developing mouse forebrain, temporally distinct waves of oligodendrocyte progenitor cells (OPCs) arise from different germinal zones and eventually populate either dorsal or ventral regions, where they present as transcriptionally and functionally equivalent cells. Despite that, developmental heterogeneity influences adult OPC responses upon demyelination. Here we show that accumulation of DNA damage due to ablation of citron-kinase or cisplatin treatment cell-autonomously disrupts OPC fate, resulting in cell death and senescence in the dorsal and ventral subsets, respectively. Such alternative fates are associated with distinct developmental origins of OPCs, and with a different activation of NRF2-mediated anti-oxidant responses. These data indicate that, upon injury, dorsal and ventral OPC subsets show functional and molecular diversity that can make them differentially vulnerable to pathological conditions associated with DNA damage.
Collapse
Affiliation(s)
- Enrica Boda
- Department of Neuroscience Rita Levi-Montalcini, University of Turin, Turin, Italy.
- Neuroscience Institute Cavalieri Ottolenghi (NICO), University of Turin, Regione Gonzole 10, IT-10043, Orbassano (Turin), Italy.
| | - Martina Lorenzati
- Department of Neuroscience Rita Levi-Montalcini, University of Turin, Turin, Italy
- Neuroscience Institute Cavalieri Ottolenghi (NICO), University of Turin, Regione Gonzole 10, IT-10043, Orbassano (Turin), Italy
| | - Roberta Parolisi
- Department of Neuroscience Rita Levi-Montalcini, University of Turin, Turin, Italy
- Neuroscience Institute Cavalieri Ottolenghi (NICO), University of Turin, Regione Gonzole 10, IT-10043, Orbassano (Turin), Italy
| | - Brian Harding
- Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania and Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Gianmarco Pallavicini
- Department of Neuroscience Rita Levi-Montalcini, University of Turin, Turin, Italy
- Neuroscience Institute Cavalieri Ottolenghi (NICO), University of Turin, Regione Gonzole 10, IT-10043, Orbassano (Turin), Italy
| | - Luca Bonfanti
- Neuroscience Institute Cavalieri Ottolenghi (NICO), University of Turin, Regione Gonzole 10, IT-10043, Orbassano (Turin), Italy
- Department of Veterinary Sciences, University of Turin, Turin, Italy
| | - Amanda Moccia
- Department of Human Genetics, University of Michigan, Ann Arbor, MI, USA
| | - Stephanie Bielas
- Department of Human Genetics, University of Michigan, Ann Arbor, MI, USA
| | - Ferdinando Di Cunto
- Department of Neuroscience Rita Levi-Montalcini, University of Turin, Turin, Italy
- Neuroscience Institute Cavalieri Ottolenghi (NICO), University of Turin, Regione Gonzole 10, IT-10043, Orbassano (Turin), Italy
| | - Annalisa Buffo
- Department of Neuroscience Rita Levi-Montalcini, University of Turin, Turin, Italy
- Neuroscience Institute Cavalieri Ottolenghi (NICO), University of Turin, Regione Gonzole 10, IT-10043, Orbassano (Turin), Italy
| |
Collapse
|
6
|
Iegiani G, Di Cunto F, Pallavicini G. Inhibiting microcephaly genes as alternative to microtubule targeting agents to treat brain tumors. Cell Death Dis 2021; 12:956. [PMID: 34663805 PMCID: PMC8523548 DOI: 10.1038/s41419-021-04259-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 09/10/2021] [Accepted: 09/24/2021] [Indexed: 01/14/2023]
Abstract
Medulloblastoma (MB) and gliomas are the most frequent high-grade brain tumors (HGBT) in children and adulthood, respectively. The general treatment for these tumors consists in surgery, followed by radiotherapy and chemotherapy. Despite the improvement in patient survival, these therapies are only partially effective, and many patients still die. In the last decades, microtubules have emerged as interesting molecular targets for HGBT, as various microtubule targeting agents (MTAs) have been developed and tested pre-clinically and clinically with encouraging results. Nevertheless, these treatments produce relevant side effects since they target microtubules in normal as well as in cancerous cells. A possible strategy to overcome this toxicity could be to target proteins that control microtubule dynamics but are required by HGBT cells much more than in normal cell types. The genes mutated in primary hereditary microcephaly (MCPH) are ubiquitously expressed in proliferating cells, but under normal conditions are selectively required during brain development, in neural progenitors. There is evidence that MB and glioma cells share molecular profiles with progenitors of cerebellar granules and of cortical radial glia cells, in which MCPH gene functions are fundamental. Moreover, several studies indicate that MCPH genes are required for HGBT expansion. Among the 25 known MCPH genes, we focus this review on KNL1, ASPM, CENPE, CITK and KIF14, which have been found to control microtubule stability during cell division. We summarize the current knowledge about the molecular basis of their interaction with microtubules. Moreover, we will discuss data that suggest these genes are promising candidates as HGBT-specific targets.
Collapse
Affiliation(s)
- Giorgia Iegiani
- Neuroscience Institute Cavalieri Ottolenghi, 10043, Orbassano, Italy
- Department of Neuroscience 'Rita Levi Montalcini', University of Turin, 10126, Turin, Italy
| | - Ferdinando Di Cunto
- Neuroscience Institute Cavalieri Ottolenghi, 10043, Orbassano, Italy
- Department of Neuroscience 'Rita Levi Montalcini', University of Turin, 10126, Turin, Italy
| | - Gianmarco Pallavicini
- Neuroscience Institute Cavalieri Ottolenghi, 10043, Orbassano, Italy.
- Department of Neuroscience 'Rita Levi Montalcini', University of Turin, 10126, Turin, Italy.
| |
Collapse
|
7
|
Pallavicini G, Gai M, Iegiani G, Berto GE, Adrait A, Couté Y, Di Cunto F. Goldberg-Shprintzen syndrome protein KIF1BP is a CITK interactor implicated in cytokinesis. J Cell Sci 2021; 134:jcs250902. [PMID: 34100550 DOI: 10.1242/jcs.250902] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 05/04/2021] [Indexed: 12/14/2022] Open
Abstract
Goldberg-Shprintzen disease (GOSHS) is a rare microcephaly syndrome accompanied by intellectual disability, dysmorphic facial features, peripheral neuropathy and Hirschsprung disease. It is associated with recessive mutations in the gene encoding kinesin family member 1-binding protein (KIF1BP, also known as KIFBP). The encoded protein regulates axon microtubules dynamics, kinesin attachment and mitochondrial biogenesis, but it is not clear how its loss could lead to microcephaly. We identified KIF1BP in the interactome of citron kinase (CITK, also known as CIT), a protein produced by the primary hereditary microcephaly 17 (MCPH17) gene. KIF1BP and CITK interact under physiological conditions in mitotic cells. Similar to CITK, KIF1BP is enriched at the midbody ring and is required for cytokinesis. The association between KIF1BP and CITK can be influenced by CITK activity, and the two proteins may antagonize each other for their midbody localization. KIF1BP knockdown decreases microtubule stability, increases KIF23 midbody levels and impairs midbody localization of KIF14, as well as of chromosome passenger complex. These data indicate that KIF1BP is a CITK interactor involved in midbody maturation and abscission, and suggest that cytokinesis failure may contribute to the microcephaly phenotype observed in GOSHS.
Collapse
Affiliation(s)
- Gianmarco Pallavicini
- Neuroscience Institute Cavalieri Ottolenghi, Turin 10123, Italy
- Department of Neuroscience 'Rita Levi Montalcini', University of Turin, Turin 10126, Italy
| | - Marta Gai
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin 10126, Italy
| | - Giorgia Iegiani
- Neuroscience Institute Cavalieri Ottolenghi, Turin 10123, Italy
- Department of Neuroscience 'Rita Levi Montalcini', University of Turin, Turin 10126, Italy
| | - Gaia Elena Berto
- Neuroscience Institute Cavalieri Ottolenghi, Turin 10123, Italy
- Department of Neuroscience 'Rita Levi Montalcini', University of Turin, Turin 10126, Italy
| | - Annie Adrait
- Univ. Grenoble Alpes, Commissariat à l'énergie atomique et aux énergies alternatives (CEA), Institut national de la santé et de la recherche médicale (INSERM), Interdisciplinary Research Institute of Grenoble (IRIG), Laboratoire Biologie à Grande Echelle (BGE), 38000 Grenoble, France
| | - Yohann Couté
- Univ. Grenoble Alpes, Commissariat à l'énergie atomique et aux énergies alternatives (CEA), Institut national de la santé et de la recherche médicale (INSERM), Interdisciplinary Research Institute of Grenoble (IRIG), Laboratoire Biologie à Grande Echelle (BGE), 38000 Grenoble, France
| | - Ferdinando Di Cunto
- Neuroscience Institute Cavalieri Ottolenghi, Turin 10123, Italy
- Department of Neuroscience 'Rita Levi Montalcini', University of Turin, Turin 10126, Italy
| |
Collapse
|
8
|
Iegiani G, Gai M, Di Cunto F, Pallavicini G. CENPE Inhibition Leads to Mitotic Catastrophe and DNA Damage in Medulloblastoma Cells. Cancers (Basel) 2021; 13:cancers13051028. [PMID: 33804489 PMCID: PMC7957796 DOI: 10.3390/cancers13051028] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 02/17/2021] [Accepted: 02/24/2021] [Indexed: 02/07/2023] Open
Abstract
Simple Summary Medulloblastoma (MB) is the most frequent brain tumor in children. The standard treatment consists in surgery, followed by radiotherapy and chemotherapy. These therapies are only partially effective, since many patients still die and those who survive suffer from neurological and endocrine disorders. Therefore, more effective therapies are needed. CENPE is a gene critical for normal proliferation and survival of neural progenitors. Since there is evidence that MB cells are very similar to neural progenitors, we hypothesized that CENPE could be an effective target for MB treatment. In MB cell lines, CENPE depletion induced defects in division and resulted in cell death. To consolidate CENPE as a target for MB treatment, we tested GSK923295, a specific inhibitor already in clinical trials for other cancer types. GSK923295 induced effects similar to CENPE depletion at low nM levels, supporting the idea that CENPE’s inhibition could be a viable strategy for MB treatment. Abstract Medulloblastoma (MB) is the most frequent brain tumor in children. The standard treatment consists in surgery, followed by radiotherapy and chemotherapy. These therapies are only partially effective since many patients still die and those who survive suffer from neurological and endocrine disorders. Therefore, more effective therapies are needed. Primary microcephaly (MCPH) is a rare disorder caused by mutations in 25 different genes. Centromere-associated protein E (CENPE) heterozygous mutations cause the MCPH13 syndrome. As for other MCPH genes, CENPE is required for normal proliferation and survival of neural progenitors. Since there is evidence that MB shares many molecular features with neural progenitors, we hypothesized that CENPE could be an effective target for MB treatment. In ONS-76 and DAOY cells, CENPE knockdown induced mitotic defects and apoptosis. Moreover, CENPE depletion induced endogenous DNA damage accumulation, activating TP53 or TP73 as well as cell death signaling pathways. To consolidate CENPE as a target for MB treatment, we tested GSK923295, an allosteric inhibitor already in clinical trial for other cancer types. GSK923295, induced effects similar to CENPE depletion with higher penetrance, at low nM levels, suggesting that CENPE’s inhibition could be a therapeutic strategy for MB treatment.
Collapse
Affiliation(s)
- Giorgia Iegiani
- Neuroscience Institute Cavalieri Ottolenghi, 10043 Turin, Italy;
- Department of Neuroscience ‘Rita Levi Montalcini’, University of Turin, 10126 Turin, Italy
| | - Marta Gai
- Department of Molecular Biotechnology and Health Sciences, University of Turin, 10126 Turin, Italy;
| | - Ferdinando Di Cunto
- Neuroscience Institute Cavalieri Ottolenghi, 10043 Turin, Italy;
- Department of Neuroscience ‘Rita Levi Montalcini’, University of Turin, 10126 Turin, Italy
- Correspondence: (F.D.C.); (G.P.)
| | - Gianmarco Pallavicini
- Neuroscience Institute Cavalieri Ottolenghi, 10043 Turin, Italy;
- Department of Neuroscience ‘Rita Levi Montalcini’, University of Turin, 10126 Turin, Italy
- Correspondence: (F.D.C.); (G.P.)
| |
Collapse
|
9
|
Zhou X, Zhi Y, Yu J, Xu D. The Yin and Yang of Autosomal Recessive Primary Microcephaly Genes: Insights from Neurogenesis and Carcinogenesis. Int J Mol Sci 2020; 21:ijms21051691. [PMID: 32121580 PMCID: PMC7084222 DOI: 10.3390/ijms21051691] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 02/23/2020] [Accepted: 02/26/2020] [Indexed: 12/26/2022] Open
Abstract
The stem cells of neurogenesis and carcinogenesis share many properties, including proliferative rate, an extensive replicative potential, the potential to generate different cell types of a given tissue, and an ability to independently migrate to a damaged area. This is also evidenced by the common molecular principles regulating key processes associated with cell division and apoptosis. Autosomal recessive primary microcephaly (MCPH) is a neurogenic mitotic disorder that is characterized by decreased brain size and mental retardation. Until now, a total of 25 genes have been identified that are known to be associated with MCPH. The inactivation (yin) of most MCPH genes leads to neurogenesis defects, while the upregulation (yang) of some MCPH genes is associated with different kinds of carcinogenesis. Here, we try to summarize the roles of MCPH genes in these two diseases and explore the underlying mechanisms, which will help us to explore new, attractive approaches to targeting tumor cells that are resistant to the current therapies.
Collapse
Affiliation(s)
- Xiaokun Zhou
- College of Biological Science and Engineering, Institute of Life Sciences, Fuzhou University, Fuzhou 350108, China; (X.Z.); (Y.Z.); (J.Y.)
| | - Yiqiang Zhi
- College of Biological Science and Engineering, Institute of Life Sciences, Fuzhou University, Fuzhou 350108, China; (X.Z.); (Y.Z.); (J.Y.)
| | - Jurui Yu
- College of Biological Science and Engineering, Institute of Life Sciences, Fuzhou University, Fuzhou 350108, China; (X.Z.); (Y.Z.); (J.Y.)
| | - Dan Xu
- College of Biological Science and Engineering, Institute of Life Sciences, Fuzhou University, Fuzhou 350108, China; (X.Z.); (Y.Z.); (J.Y.)
- Fujian Key Laboratory of Molecular Neurology, Institute of Neuroscience, Fujian Medical University, Fuzhou 350005, China
- Correspondence: ; Tel.: +86-17085937559
| |
Collapse
|
10
|
CITK Loss Inhibits Growth of Group 3 and Group 4 Medulloblastoma Cells and Sensitizes Them to DNA-Damaging Agents. Cancers (Basel) 2020; 12:cancers12030542. [PMID: 32111106 PMCID: PMC7139701 DOI: 10.3390/cancers12030542] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 02/15/2020] [Accepted: 02/23/2020] [Indexed: 02/06/2023] Open
Abstract
Medulloblastoma (MB) is the most common malignant brain tumor in children, and it is classified into four biological subgroups: WNT, Sonic Hedgehog (SHH), Group 3 and Group 4. The current treatment is surgery, followed by irradiation and chemotherapy. Unfortunately, these therapies are only partially effective. Citron kinase protein (CITK) has been proposed as a promising target for SHH MB, whose inactivation leads to DNA damage and apoptosis. D283 and D341 cell lines (Group 3/Group 4 MB) were silenced with established siRNA sequences against CITK, to assess the direct effects of its loss. Next, D283, D341, ONS-76 and DAOY cells were treated with ionizing radiation (IR) or cisplatin in combination with CITK knockdown. CITK depletion impaired proliferation and induced cytokinesis failure and apoptosis of G3/G4 MB cell lines. Furthermore, CITK knockdown produced an accumulation of DNA damage, with reduced RAD51 nuclear levels. Association of IR or cisplatin with CITK depletion strongly impaired the growth potential of all tested MB cells. These results indicate that CITK inactivation could prevent the expansion of G3/G4 MB and increase their sensitivity to DNA-damaging agents, by impairing homologous recombination. We suggest that CITK inhibition could be broadly associated with IR and adjuvant therapy in MB treatment.
Collapse
|
11
|
Citron Rho-Interacting Serine/Threonine Kinase Promotes HIF1a-CypA Signaling and Growth of Human Pancreatic Adenocarcinoma. BIOMED RESEARCH INTERNATIONAL 2020; 2020:9210891. [PMID: 32185224 PMCID: PMC7060418 DOI: 10.1155/2020/9210891] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 01/19/2020] [Accepted: 01/23/2020] [Indexed: 01/11/2023]
Abstract
In human pancreatic ductal adenocarcinoma (PDAC), the cyclophilin A (CypA) is overexpressed and promotes the development of PDAC. However, the mechanism underlying cyclophilin A expression remains elusive. Here, we reported that the citron Rho-interacting serine/threonine kinase (CIT) promotes the HIF1a-CypA signaling and growth of PDAC cells. CIT expression was higher in PDAC cells compared with the normal epithelial cells, and clinical data showed that CIT was overexpressed in PDAC tissues and high expression of CIT predicted poor overall and disease-free survival. In PDAC cells, knockdown of CIT expression repressed the rate of proliferation and capacity of colony formation, which were accomplished with an increased percentage of apoptotic cells and cell cycle arrest. The knockdown of CIT in PDAC cells reduced the expression of CypA while overexpression of CIT promoted the expression of CypA. We observed that the effects of CIT on the expression of CypA relied on the transcriptional factor HIF1a, which was previously reported to transcriptionally activate the expression of CypA in PDAC cells. Furthermore, the effects of CIT on apoptosis, cell cycle, proliferation, and colony formation of PDAC cells relied on its role in the regulation of CypA expression. Collectively, our data showed that CIT promoted the activation of HIF1-CypA signaling and enhanced the growth of PDAC cells.
Collapse
|
12
|
Smirnov A, Cappello A, Lena AM, Anemona L, Mauriello A, Di Daniele N, Annicchiarico-Petruzzelli M, Melino G, Candi E. ZNF185 is a p53 target gene following DNA damage. Aging (Albany NY) 2019; 10:3308-3326. [PMID: 30446632 PMCID: PMC6286825 DOI: 10.18632/aging.101639] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Accepted: 11/01/2018] [Indexed: 12/17/2022]
Abstract
The transcription factor p53 is a key player in the tumour suppressive DNA damage response and a growing number of target genes involved in these pathways has been identified. p53 has been shown to be implicated in controlling cell motility and its mutant form enhances metastasis by loss of cell directionality, but the p53 role in this context has not yet being investigated. Here, we report that ZNF185, an actin cytoskeleton-associated protein from LIM-family of Zn-finger proteins, is induced following DNA-damage. ChIP-seq analysis, chromatin crosslinking immune-precipitation experiments and luciferase assays demonstrate that ZNF185 is a bona fide p53 target gene. Upon genotoxic stress, caused by DNA-damaging drug etoposide and UVB irradiation, ZNF185 expression is up-regulated and in etoposide-treated cells, ZNF185 depletion does not affect cell proliferation and apoptosis, but interferes with actin cytoskeleton remodelling and cell polarization. Bioinformatic analysis of different types of epithelial cancers from both TCGA and GTEx databases showed a significant decrease in ZNF185 mRNA level compared to normal tissues. These findings are confirmed by tissue micro-array IHC staining. Our data highlight the involvement of ZNF185 and cytoskeleton changes in p53-mediated cellular response to genotoxic stress and indicate ZNF185 as potential biomarker for epithelial cancer diagnosis.
Collapse
Affiliation(s)
- Artem Smirnov
- Department of Experimental Medicine, TOR, University of Rome "Tor Vergata", Rome 00133, Italy
| | - Angela Cappello
- Department of Experimental Medicine, TOR, University of Rome "Tor Vergata", Rome 00133, Italy
| | - Anna Maria Lena
- Department of Experimental Medicine, TOR, University of Rome "Tor Vergata", Rome 00133, Italy
| | - Lucia Anemona
- Department of Experimental Medicine, TOR, University of Rome "Tor Vergata", Rome 00133, Italy
| | - Alessandro Mauriello
- Department of Experimental Medicine, TOR, University of Rome "Tor Vergata", Rome 00133, Italy
| | - Nicola Di Daniele
- Department of Systems Medicine, University of Rome "Tor Vergata", Rome 00133, Italy
| | | | - Gerry Melino
- Department of Experimental Medicine, TOR, University of Rome "Tor Vergata", Rome 00133, Italy.,MRC-Toxicology Unit, University of Cambridge, Cambridge, UK
| | - Eleonora Candi
- Department of Experimental Medicine, TOR, University of Rome "Tor Vergata", Rome 00133, Italy.,Istituto Dermopatico dell'Immacolata-IRCCS, Rome 00163, Italy
| |
Collapse
|
13
|
Magliozzi R, Carrero ZI, Low TY, Yuniati L, Valdes-Quezada C, Kruiswijk F, van Wijk K, Heck AJR, Jackson CL, Guardavaccaro D. Inheritance of the Golgi Apparatus and Cytokinesis Are Controlled by Degradation of GBF1. Cell Rep 2019; 23:3381-3391.e4. [PMID: 29898406 DOI: 10.1016/j.celrep.2018.05.031] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Revised: 04/05/2018] [Accepted: 05/10/2018] [Indexed: 11/27/2022] Open
Abstract
Although much is known about how chromosome segregation is coupled to cell division, how intracellular organelles partition during mitotic division is poorly understood. We report that the phosphorylation-dependent degradation of the ARFGEF GBF1 regulates organelle trafficking during cell division. We show that, in mitosis, GBF1 is phosphorylated on Ser292 and Ser297 by casein kinase-2 allowing recognition by the F-box protein βTrCP. GBF1 interaction with βTrCP recruits GBF1 to the SCFβTrCP ubiquitin ligase complex, triggering its degradation. Phosphorylation and degradation of GBF1 occur along microtubules at the intercellular bridge of telophase cells and are required for Golgi membrane positioning and postmitotic Golgi reformation. Indeed, expression of a non-degradable GBF1 mutant inhibits the transport of the Golgi cluster adjacent to the midbody toward the Golgi twin positioned next to the centrosome and results in defective Golgi reassembly and cytokinesis failure. These findings define a mechanism that controls postmitotic Golgi reassembly and inheritance.
Collapse
Affiliation(s)
- Roberto Magliozzi
- Hubrecht Institute-KNAW and University Medical Center Utrecht, Uppsalalaan 8, 3584 Utrecht, the Netherlands
| | - Zunamys I Carrero
- Hubrecht Institute-KNAW and University Medical Center Utrecht, Uppsalalaan 8, 3584 Utrecht, the Netherlands
| | - Teck Yew Low
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Padualaan 8, 3584 Utrecht, the Netherlands; The Netherlands Proteomics Center, Padualaan 8, 3584 Utrecht, the Netherlands
| | - Laurensia Yuniati
- Hubrecht Institute-KNAW and University Medical Center Utrecht, Uppsalalaan 8, 3584 Utrecht, the Netherlands
| | - Christian Valdes-Quezada
- Oncode Institute, Hubrecht Institute-KNAW and University Medical Center Utrecht, Uppsalalaan 8, 3584 Utrecht, the Netherlands
| | - Flore Kruiswijk
- Hubrecht Institute-KNAW and University Medical Center Utrecht, Uppsalalaan 8, 3584 Utrecht, the Netherlands
| | - Koen van Wijk
- Hubrecht Institute-KNAW and University Medical Center Utrecht, Uppsalalaan 8, 3584 Utrecht, the Netherlands
| | - Albert J R Heck
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Padualaan 8, 3584 Utrecht, the Netherlands; The Netherlands Proteomics Center, Padualaan 8, 3584 Utrecht, the Netherlands
| | - Catherine L Jackson
- Membrane Dynamics and Intracellular Trafficking, Institut Jacques Monod, CNRS, UMR 7592, Université Paris Diderot, Sorbonne Paris Cité, 75013 Paris, France
| | - Daniele Guardavaccaro
- Hubrecht Institute-KNAW and University Medical Center Utrecht, Uppsalalaan 8, 3584 Utrecht, the Netherlands.
| |
Collapse
|
14
|
D'Amore C, Salizzato V, Borgo C, Cesaro L, Pinna LA, Salvi M. A Journey through the Cytoskeleton with Protein Kinase CK2. Curr Protein Pept Sci 2019; 20:547-562. [PMID: 30659536 DOI: 10.2174/1389203720666190119124846] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Revised: 12/21/2018] [Accepted: 01/09/2019] [Indexed: 01/15/2023]
Abstract
Substrate pleiotropicity, a very acidic phosphorylation consensus sequence, and an apparent uncontrolled activity, are the main features of CK2, a Ser/Thr protein kinase that is required for a plethora of cell functions. Not surprisingly, CK2 appears to affect cytoskeletal structures and correlated functions such as cell shape, mechanical integrity, cell movement and division. This review outlines our current knowledge of how CK2 regulates cytoskeletal structures, and discusses involved pathways and molecular mechanisms.
Collapse
Affiliation(s)
- Claudio D'Amore
- Department of Biomedical Sciences, University of Padova, Via U. Bassi 58/B, Padova, Italy
| | - Valentina Salizzato
- Department of Biomedical Sciences, University of Padova, Via U. Bassi 58/B, Padova, Italy.,CNR Institute of Neurosciences, Via U. Bassi 58/B, Padova, Italy
| | - Christian Borgo
- Department of Biomedical Sciences, University of Padova, Via U. Bassi 58/B, Padova, Italy
| | - Luca Cesaro
- Department of Biomedical Sciences, University of Padova, Via U. Bassi 58/B, Padova, Italy
| | - Lorenzo A Pinna
- Department of Biomedical Sciences, University of Padova, Via U. Bassi 58/B, Padova, Italy.,CNR Institute of Neurosciences, Via U. Bassi 58/B, Padova, Italy
| | - Mauro Salvi
- Department of Biomedical Sciences, University of Padova, Via U. Bassi 58/B, Padova, Italy
| |
Collapse
|
15
|
El-Amine N, Carim SC, Wernike D, Hickson GRX. Rho-dependent control of the Citron kinase, Sticky, drives midbody ring maturation. Mol Biol Cell 2019; 30:2185-2204. [PMID: 31166845 PMCID: PMC6743463 DOI: 10.1091/mbc.e19-04-0194] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Rho-dependent proteins control assembly of the cytokinetic contractile ring, yet it remains unclear how those proteins guide ring closure and how they promote subsequent formation of a stable midbody ring. Citron kinase is one important component required for midbody ring formation but its mechanisms of action and relationship with Rho are controversial. Here, we conduct a structure-function analysis of the Drosophila Citron kinase, Sticky, in Schneider's S2 cells. We define two separable and redundant RhoGEF/Pebble-dependent inputs into Sticky recruitment to the nascent midbody ring and show that each input is subsequently required for retention at, and for the integrity of, the mature midbody ring. The first input is via an actomyosin-independent interaction between Sticky and Anillin, a key scaffold also required for midbody ring formation. The second input requires the Rho-binding domain of Sticky, whose boundaries we have defined. Collectively, these results show how midbody ring biogenesis depends on the coordinated actions of Sticky, Anillin, and Rho.
Collapse
Affiliation(s)
- Nour El-Amine
- Centre de Cancérologie Charles Bruneau, Centre Hospitalier Universitaire Sainte-Justine Centre de Recherche, Montréal, QC H3T 1C5, Canada.,Département de Pathologie et Biologie Cellulaire, Université de Montréal, Montréal, QC H3C 3J7, Canada
| | - Sabrya C Carim
- Centre de Cancérologie Charles Bruneau, Centre Hospitalier Universitaire Sainte-Justine Centre de Recherche, Montréal, QC H3T 1C5, Canada
| | - Denise Wernike
- Centre de Cancérologie Charles Bruneau, Centre Hospitalier Universitaire Sainte-Justine Centre de Recherche, Montréal, QC H3T 1C5, Canada
| | - Gilles R X Hickson
- Centre de Cancérologie Charles Bruneau, Centre Hospitalier Universitaire Sainte-Justine Centre de Recherche, Montréal, QC H3T 1C5, Canada.,Département de Pathologie et Biologie Cellulaire, Université de Montréal, Montréal, QC H3C 3J7, Canada
| |
Collapse
|
16
|
Pallavicini G, Berto GE, Di Cunto F. Precision Revisited: Targeting Microcephaly Kinases in Brain Tumors. Int J Mol Sci 2019; 20:ijms20092098. [PMID: 31035417 PMCID: PMC6539168 DOI: 10.3390/ijms20092098] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 04/24/2019] [Accepted: 04/26/2019] [Indexed: 12/18/2022] Open
Abstract
Glioblastoma multiforme and medulloblastoma are the most frequent high-grade brain tumors in adults and children, respectively. Standard therapies for these cancers are mainly based on surgical resection, radiotherapy, and chemotherapy. However, intrinsic or acquired resistance to treatment occurs almost invariably in the first case, and side effects are unacceptable in the second. Therefore, the development of new, effective drugs is a very important unmet medical need. A critical requirement for developing such agents is to identify druggable targets required for the proliferation or survival of tumor cells, but not of other cell types. Under this perspective, genes mutated in congenital microcephaly represent interesting candidates. Congenital microcephaly comprises a heterogeneous group of disorders in which brain volume is reduced, in the absence or presence of variable syndromic features. Genetic studies have clarified that most microcephaly genes encode ubiquitous proteins involved in mitosis and in maintenance of genomic stability, but the effects of their inactivation are particularly strong in neural progenitors. It is therefore conceivable that the inhibition of the function of these genes may specifically affect the proliferation and survival of brain tumor cells. Microcephaly genes encode for a few kinases, including CITK, PLK4, AKT3, DYRK1A, and TRIO. In this review, we summarize the evidence indicating that the inhibition of these molecules could exert beneficial effects on different aspects of brain cancer treatment.
Collapse
Affiliation(s)
- Gianmarco Pallavicini
- Neuroscience Institute Cavalieri Ottolenghi, 10126 Turin, Italy.
- Department of Neurosciences, University of Turin, 10126 Turin, Italy.
- Department of Molecular Biotechnology and Health Sciences, University of Turin, 10126 Turin, Italy.
| | - Gaia E Berto
- Neuroscience Institute Cavalieri Ottolenghi, 10126 Turin, Italy.
- Department of Neurosciences, University of Turin, 10126 Turin, Italy.
| | - Ferdinando Di Cunto
- Neuroscience Institute Cavalieri Ottolenghi, 10126 Turin, Italy.
- Department of Neurosciences, University of Turin, 10126 Turin, Italy.
- Neuroscience Institute of Turin (NIT), 10126 Turin, Italy.
| |
Collapse
|
17
|
Sahin I, Kawano Y, Sklavenitis-Pistofidis R, Moschetta M, Mishima Y, Manier S, Sacco A, Carrasco R, Fonseca R, Roccaro AM, Witzig T, Ghobrial IM. Citron Rho-interacting kinase silencing causes cytokinesis failure and reduces tumor growth in multiple myeloma. Blood Adv 2019; 3:995-1002. [PMID: 30940634 PMCID: PMC6457230 DOI: 10.1182/bloodadvances.2018028456] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Accepted: 02/21/2019] [Indexed: 11/20/2022] Open
Abstract
Citron Rho-interacting serine/threonine kinase (CIT) is a serine/threonine kinase that acts as a key component of the midbody and is essential for cytokinesis. CIT has been reported to be highly expressed in some tumor tissues and to play a role in cancer proliferation; however, the significance of CIT has not been investigated in multiple myeloma (MM). Here, we identified, by protein microarray and immunohistochemistry, that CIT is 1 of the upregulated proteins in the plasma cells of MM patients compared with healthy controls. Analysis of a gene expression profile data set showed that MM patients with high CIT gene expression had significantly worse overall survival compared with MM patients with low CIT gene expression. CIT silencing in MM cell lines induced cytokinesis failure and resulted in decreased MM cell proliferation in vitro and in vivo. TP53 expression was found to be an independent predictor of CIT dependency, with low-TP53 cell lines exhibiting a strong dependency on CIT. This study provides the rationale for CIT being a potential therapeutic target in MM in future trials.
Collapse
Affiliation(s)
- Ilyas Sahin
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
- Division of Hematology-Oncology, Lifespan Cancer Institute, The Warren Alpert Medical School of Brown University, Providence, RI
| | - Yawara Kawano
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
- Department of Hematology, Kumamoto University Hospital, Kumamoto, Japan
| | | | - Michele Moschetta
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - Yuji Mishima
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - Salomon Manier
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - Antonio Sacco
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
- ASST Spedali Civili di Brescia Clinical Research Development and Phase I Unit-CREA Laboratory, Brescia, Italy
| | - Ruben Carrasco
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - Rafael Fonseca
- Division of Hematology, Mayo Clinic, Scottsdale, AZ; and
| | - Aldo M Roccaro
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
- ASST Spedali Civili di Brescia Clinical Research Development and Phase I Unit-CREA Laboratory, Brescia, Italy
| | | | - Irene M Ghobrial
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| |
Collapse
|
18
|
Calorio C, Gavello D, Guarina L, Salio C, Sassoè-Pognetto M, Riganti C, Bianchi FT, Hofer NT, Tuluc P, Obermair GJ, Defilippi P, Balzac F, Turco E, Bett GC, Rasmusson RL, Carbone E. Impaired chromaffin cell excitability and exocytosis in autistic Timothy syndrome TS2-neo mouse rescued by L-type calcium channel blockers. J Physiol 2019; 597:1705-1733. [PMID: 30629744 DOI: 10.1113/jp277487] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Accepted: 12/19/2018] [Indexed: 12/30/2022] Open
Abstract
KEY POINTS Tymothy syndrome (TS) is a multisystem disorder featuring cardiac arrhythmias, autism and adrenal gland dysfunction that originates from a de novo point mutation in the gene encoding the Cav1.2 (CACNA1C) L-type channel. To study the role of Cav1.2 channel signals in autism, the autistic TS2-neo mouse has been generated bearing the G406R point-mutation associated with TS type-2. Using heterozygous TS2-neo mice, we report that the G406R mutation reduces the rate of inactivation and shifts leftward the activation and inactivation of L-type channels, causing marked increase of resting Ca2+ influx ('window' Ca2+ current). The increased 'window current' causes marked reduction of NaV channel density, switches normal tonic firing to abnormal burst firing, reduces mitochondrial metabolism, induces cell swelling and decreases catecholamine release. Overnight incubations with nifedipine rescue NaV channel density, normal firing and the quantity of catecholamine released. We provide evidence that chromaffin cell malfunction derives from altered Cav1.2 channel gating. ABSTRACT L-type voltage-gated calcium (Cav1) channels have a key role in long-term synaptic plasticity, sensory transduction, muscle contraction and hormone release. A point mutation in the gene encoding Cav1.2 (CACNA1C) causes Tymothy syndrome (TS), a multisystem disorder featuring cardiac arrhythmias, autism spectrum disorder (ASD) and adrenal gland dysfunction. In the more severe type-2 form (TS2), the missense mutation G406R is on exon 8 coding for the IS6-helix of the Cav1.2 channel. The mutation causes reduced inactivation and induces autism. How this occurs and how Cav1.2 gating-changes alter cell excitability, neuronal firing and hormone release on a molecular basis is still largely unknown. Here, using the TS2-neo mouse model of TS we show that the G406R mutation altered excitability and reduced secretory activity in adrenal chromaffin cells (CCs). Specifically, the TS2 mutation reduced the rate of voltage-dependent inactivation and shifted leftward the activation and steady-state inactivation of L-type channels. This markedly increased the resting 'window' Ca2+ current that caused an increased percentage of CCs undergoing abnormal action potential (AP) burst firing, cell swelling, reduced mitochondrial metabolism and decreased catecholamine release. The increased 'window' Ca2+ current caused also decreased NaV channel density and increased steady-state inactivation, which contributed to the increased abnormal burst firing. Overnight incubation with the L-type channel blocker nifedipine rescued the normal AP firing of CCs, the density of functioning NaV channels and their steady-state inactivation. We provide evidence that CC malfunction derives from the altered Cav1.2 channel gating and that dihydropyridines are potential therapeutics for ASD.
Collapse
Affiliation(s)
- Chiara Calorio
- Department of Drug Science, NIS Centre, University of Torino, Torino, Italy
| | - Daniela Gavello
- Department of Drug Science, NIS Centre, University of Torino, Torino, Italy
| | - Laura Guarina
- Department of Drug Science, NIS Centre, University of Torino, Torino, Italy
| | - Chiara Salio
- Department of Veterinary Sciences, University of Torino, Torino, Italy
| | - Marco Sassoè-Pognetto
- Department of Neuroscience Rita Levi Montalcini, University of Torino, Torino, Italy
| | - Chiara Riganti
- Department of Oncology, University of Torino, Torino, Italy
| | | | - Nadja T Hofer
- Department of Pharmacology and Toxicology, Center for Molecular Biosciences, University of Innsbruck, Innsbruck, Austria
| | - Petronel Tuluc
- Department of Pharmacology and Toxicology, Center for Molecular Biosciences, University of Innsbruck, Innsbruck, Austria
| | - Gerald J Obermair
- Department of Physiology & Medical Physics, Medical University of Innsbruck, Innsbruck, Austria
| | - Paola Defilippi
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Fiorella Balzac
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Emilia Turco
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Glenna C Bett
- Department of Physiology & Biophysics, State University of New York, Buffalo, NY, USA
| | - Randall L Rasmusson
- Department of Physiology & Biophysics, State University of New York, Buffalo, NY, USA
| | - Emilio Carbone
- Department of Drug Science, NIS Centre, University of Torino, Torino, Italy
| |
Collapse
|
19
|
Bianchi FT, Berto GE, Di Cunto F. Impact of DNA repair and stability defects on cortical development. Cell Mol Life Sci 2018; 75:3963-3976. [PMID: 30116853 PMCID: PMC11105354 DOI: 10.1007/s00018-018-2900-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Revised: 07/16/2018] [Accepted: 08/08/2018] [Indexed: 02/07/2023]
Abstract
Maintenance of genome stability is a crucial cellular function for normal mammalian development and physiology. However, despite the general relevance of this process, genome stability alteration due to genetic or non-genetic conditions has a particularly profound impact on the developing cerebral cortex. In this review, we will analyze the main pathways involved in maintenance of genome stability, the consequences of their alterations with regard to central nervous system development, as well as the possible molecular and cellular basis of this specificity.
Collapse
Affiliation(s)
- Federico T Bianchi
- Neuroscience Institute Cavalieri Ottolenghi, Turin, Italy.
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy.
| | - Gaia E Berto
- Neuroscience Institute Cavalieri Ottolenghi, Turin, Italy
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Ferdinando Di Cunto
- Neuroscience Institute Cavalieri Ottolenghi, Turin, Italy
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
- Department of Neuroscience, University of Turin, Turin, Italy
| |
Collapse
|
20
|
Davies T, Kim HX, Romano Spica N, Lesea-Pringle BJ, Dumont J, Shirasu-Hiza M, Canman JC. Cell-intrinsic and -extrinsic mechanisms promote cell-type-specific cytokinetic diversity. eLife 2018; 7:36204. [PMID: 30028292 PMCID: PMC6054530 DOI: 10.7554/elife.36204] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2018] [Accepted: 06/10/2018] [Indexed: 01/05/2023] Open
Abstract
Cytokinesis, the physical division of one cell into two, is powered by constriction of an actomyosin contractile ring. It has long been assumed that all animal cells divide by a similar molecular mechanism, but growing evidence suggests that cytokinetic regulation in individual cell types has more variation than previously realized. In the four-cell Caenorhabditis elegans embryo, each blastomere has a distinct cell fate, specified by conserved pathways. Using fast-acting temperature-sensitive mutants and acute drug treatment, we identified cell-type-specific variation in the cytokinetic requirement for a robust forminCYK-1-dependent filamentous-actin (F-actin) cytoskeleton. In one cell (P2), this cytokinetic variation is cell-intrinsically regulated, whereas in another cell (EMS) this variation is cell-extrinsically regulated, dependent on both SrcSRC-1 signaling and direct contact with its neighbor cell, P2. Thus, both cell-intrinsic and -extrinsic mechanisms control cytokinetic variation in individual cell types and can protect against division failure when the contractile ring is weakened. The successful division of one cell into two is essential for all organisms to live, grow and reproduce. For an animal cell, the nucleus – the compartment containing the genetic material – must divide before the surrounding material. The rest of the cell, called the cytoplasm, physically separates later in a process known as cytokinesis. Cytokinesis in animal cells is driven by the formation of a ring in the middle of the dividing cell. The ring is composed of myosin motor proteins and filaments made of a protein called actin. The movements of the motor proteins along the filaments cause the ring to contract and tighten. This pulls the cell membrane inward and physically pinches the cell into two. For a long time, the mechanism of cytokinesis was assumed to be same across different types of animal cell, but later evidence suggested otherwise. For example, in liver, heat and bone cells, cytokinesis naturally fails during development to create cells with two or more nuclei. If a similar ‘failure’ happened in other cell types, it could lead to diseases such as cancers or blood disorders. This raised the question: what are the molecular mechanisms that allow cytokinesis to happen differently in different cell types? Davies et al. investigated this question using embryos of the worm Caenorhabditis elegans at a stage in their development when they consist of just four cells. The proteins forming the contractile ring in this worm are the same as those in humans. However, in the worm, the contractile ring can easily be damaged using chemical inhibitors or by mutating the genes that encode its proteins. Davies et al. show that when the contractile ring was damaged, two of the four cells in the worm embryo still divided successfully. This result indicates the existence of new mechanisms to divide the cytoplasm that allow division even with a weak contractile ring. In a further experiment, the embryos were dissected to isolate each of the four cells. Davies et al. saw that one of the two dividing cells could still divide on its own, while the other cell could not. This shows that this new method of cytokinesis is regulated both by factors inherent to the dividing cell and by external signals from other cells. Moreover, one of these extrinsic signals was found to be a signaling protein that had previously been implicated in human cancers. Future work will determine if these variations in cytokinesis between the different cell types found in the worm apply to humans too; and, more importantly from a therapeutic standpoint, if these new mechanisms exist in human cancers.
Collapse
Affiliation(s)
- Tim Davies
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, United States
| | - Han X Kim
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, United States.,Department of Genetics and Development, Columbia University Medical Center, New York, United States
| | - Natalia Romano Spica
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, United States
| | - Benjamin J Lesea-Pringle
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, United States
| | - Julien Dumont
- Institut Jacques Monod, CNRS UMR 7592, Université Paris Diderot, Paris, France
| | - Mimi Shirasu-Hiza
- Department of Genetics and Development, Columbia University Medical Center, New York, United States
| | - Julie C Canman
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, United States
| |
Collapse
|
21
|
Pallavicini G, Sgrò F, Garello F, Falcone M, Bitonto V, Berto GE, Bianchi FT, Gai M, Chiotto AM, Filippi M, Cutrin JC, Ala U, Terreno E, Turco E, Cunto FD. Inactivation of Citron Kinase Inhibits Medulloblastoma Progression by Inducing Apoptosis and Cell Senescence. Cancer Res 2018; 78:4599-4612. [DOI: 10.1158/0008-5472.can-17-4060] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2017] [Revised: 05/01/2018] [Accepted: 06/07/2018] [Indexed: 11/16/2022]
|
22
|
McKenzie C, D'Avino PP. Investigating cytokinesis failure as a strategy in cancer therapy. Oncotarget 2018; 7:87323-87341. [PMID: 27895316 PMCID: PMC5349991 DOI: 10.18632/oncotarget.13556] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Accepted: 11/02/2016] [Indexed: 12/12/2022] Open
Abstract
Effective therapeutics exploit common characteristics shared amongst cancers. As many cancers present chromosomal instability (CIN), one possible approach to treat these cancers could be to increase their CIN above a threshold that would affect their viability. Here, we investigated whether causing polyploidy by cytokinesis failure could represent a useful approach. We show that cytokinesis failure caused by depletion of Citron kinase (CIT-K) dramatically decreased cell proliferation in breast, cervical and colorectal cancer cells. CIT-K depletion activated the Hippo tumor suppressor pathway in normal, but not in cancer cells, indicating that cancer cells have evolved mechanisms to bypass this control. CIT-K depleted cancer cells died via apoptosis in a caspase 7 dependent manner and, consistent with this, p53-deficient HCT116 colon carcinoma cells failed to induce apoptosis after cytokinesis failure. However, other p53-mutated cancer cells were able to initiate apoptosis, indicating that cytokinesis failure can trigger apoptosis through a p53-independent mechanism. Finally, we found that actively dividing and, in some cases, polyploid cancer cells were more susceptible to CIT-K depletion. In sum, our findings indicate that inducing cytokinesis failure could be a promising anti-cancer therapeutic approach for a wide range of cancers, especially those characterized by fast cell proliferation and polyploidy.
Collapse
Affiliation(s)
- Callum McKenzie
- Department of Pathology, University of Cambridge, Cambridge, CB2 1QP, UK
| | - Pier Paolo D'Avino
- Department of Pathology, University of Cambridge, Cambridge, CB2 1QP, UK
| |
Collapse
|
23
|
Dema A, Macaluso F, Sgrò F, Berto GE, Bianchi FT, Chiotto AA, Pallavicini G, Di Cunto F, Gai M. Citron kinase-dependent F-actin maintenance at midbody secondary ingression sites mediates abscission. J Cell Sci 2018; 131:jcs.209080. [DOI: 10.1242/jcs.209080] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2017] [Accepted: 03/16/2018] [Indexed: 01/22/2023] Open
Abstract
Abscission is the final step of cytokinesis whereby the intercellular bridge (ICB) linking the two daughter cells is cut. The ICB contains a structure called the midbody, required for the recruitment and organization of the abscission machinery. Final midbody severing is mediated by formation of secondary midbody ingression sites, where ESCRT III component CHMP4B is recruited and may mediate membrane fusion. It is presently unknown how cytoskeletal elements cooperate with CHMP4B to mediate abscission. In this report, we show that F-actin is associated with midbody secondary sites and is necessary for abscission. F-actin localization at secondary sites depends on the activity of RhoA and on the abscission regulator CITK. CITK depletion accelerates F-actin loss at the midbody and cytokinesis defects produced by CITK loss are reverted by restoring actin polymerization. Conversely, midbody hyperstabilization produced by CITK and ANLN overexpression is reverted by actin depolymerization. CITK is required for F-actin and ANLN localization at the abscission sites, as well as for CHMP4B recruitment. These results indicate that control of actin dynamics downstream of CITK prepares abscission site for final cut.
Collapse
Affiliation(s)
- Alessandro Dema
- Dept. of Molecular Biotechnology and Health Sciences, University of Turin, Italy
- FMP-Berlin Campus Berlin-Buch, Robert-Roessle-Str. 10, 13125 Berlin, Germany
| | - Francesca Macaluso
- Dept. of Molecular Biotechnology and Health Sciences, University of Turin, Italy
| | - Francesco Sgrò
- Dept. of Molecular Biotechnology and Health Sciences, University of Turin, Italy
| | - Gaia E. Berto
- Dept. of Molecular Biotechnology and Health Sciences, University of Turin, Italy
- Neuroscience Institute Cavalieri Ottolenghi, Turin, Italy
| | - Federico T. Bianchi
- Dept. of Molecular Biotechnology and Health Sciences, University of Turin, Italy
- Neuroscience Institute Cavalieri Ottolenghi, Turin, Italy
| | - Alessandra A. Chiotto
- Dept. of Molecular Biotechnology and Health Sciences, University of Turin, Italy
- Neuroscience Institute Cavalieri Ottolenghi, Turin, Italy
| | - Gianmarco Pallavicini
- Dept. of Molecular Biotechnology and Health Sciences, University of Turin, Italy
- Neuroscience Institute Cavalieri Ottolenghi, Turin, Italy
| | - Ferdinando Di Cunto
- Dept. of Molecular Biotechnology and Health Sciences, University of Turin, Italy
- Neuroscience Institute Cavalieri Ottolenghi, Turin, Italy
- Neuroscience Institute of Turin (NIT), Turin, Italy
| | - Marta Gai
- Dept. of Molecular Biotechnology and Health Sciences, University of Turin, Italy
| |
Collapse
|
24
|
Bianchi FT, Gai M, Berto GE, Di Cunto F. Of rings and spines: The multiple facets of Citron proteins in neural development. Small GTPases 2017; 11:122-130. [PMID: 29185861 PMCID: PMC7053930 DOI: 10.1080/21541248.2017.1374325] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022] Open
Abstract
The Citron protein was originally identified for its capability to specifically bind the active form of RhoA small GTPase, leading to the simplistic hypothesis that it may work as a RhoA downstream effector in actin remodeling. More than two decades later, a much more complex picture has emerged. In particular, it has become clear that in animals, and especially in mammals, the functions of the Citron gene (CIT) are intimately linked to many aspects of central nervous system (CNS) development and function, although the gene is broadly expressed. More specifically, CIT encodes two main isoforms, Citron-kinase (CIT-K) and Citron-N (CIT-N), characterized by complementary expression pattern and different functions. Moreover, in many of their activities, CIT proteins act more as upstream regulators than as downstream effectors of RhoA. Finally it has been found that, besides working through actin, CIT proteins have many crucial functional interactions with the microtubule cytoskeleton and may directly affect genome stability. In this review, we will summarize these advances and illustrate their actual or potential relevance for CNS diseases, including microcephaly and psychiatric disorders.
Collapse
Affiliation(s)
- Federico T Bianchi
- Neuroscience Institute Cavalieri Ottolenghi, Regione Golzole 10, Orbassano, TO, Italy.,Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Marta Gai
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Gaia E Berto
- Neuroscience Institute Cavalieri Ottolenghi, Regione Golzole 10, Orbassano, TO, Italy.,Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Ferdinando Di Cunto
- Neuroscience Institute Cavalieri Ottolenghi, Regione Golzole 10, Orbassano, TO, Italy.,Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| |
Collapse
|
25
|
Bianchi FT, Tocco C, Pallavicini G, Liu Y, Vernì F, Merigliano C, Bonaccorsi S, El-Assawy N, Priano L, Gai M, Berto GE, Chiotto AMA, Sgrò F, Caramello A, Tasca L, Ala U, Neri F, Oliviero S, Mauro A, Geley S, Gatti M, Di Cunto F. Citron Kinase Deficiency Leads to Chromosomal Instability and TP53-Sensitive Microcephaly. Cell Rep 2017; 18:1674-1686. [PMID: 28199840 PMCID: PMC5318669 DOI: 10.1016/j.celrep.2017.01.054] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Revised: 12/16/2016] [Accepted: 01/22/2017] [Indexed: 11/16/2022] Open
Abstract
Mutations in citron (CIT), leading to loss or inactivation of the citron kinase protein (CITK), cause primary microcephaly in humans and rodents, associated with cytokinesis failure and apoptosis in neural progenitors. We show that CITK loss induces DNA damage accumulation and chromosomal instability in both mammals and Drosophila. CITK-deficient cells display "spontaneous" DNA damage, increased sensitivity to ionizing radiation, and defective recovery from radiation-induced DNA lesions. In CITK-deficient cells, DNA double-strand breaks increase independently of cytokinesis failure. Recruitment of RAD51 to DNA damage foci is compromised by CITK loss, and CITK physically interacts with RAD51, suggesting an involvement of CITK in homologous recombination. Consistent with this scenario, in doubly CitK and Trp53 mutant mice, neural progenitor cell death is dramatically reduced; moreover, clinical and neuroanatomical phenotypes are remarkably improved. Our results underscore a crucial role of CIT in the maintenance of genomic integrity during brain development.
Collapse
Affiliation(s)
- Federico Tommaso Bianchi
- Department of Molecular Biotechnology and Health Sciences, University of Turin, 10126 Turin, Italy; Neuroscience Institute Cavalieri Ottolenghi, University of Turin, 10043 Orbassano (TO), Italy.
| | - Chiara Tocco
- Department of Molecular Biotechnology and Health Sciences, University of Turin, 10126 Turin, Italy
| | - Gianmarco Pallavicini
- Department of Molecular Biotechnology and Health Sciences, University of Turin, 10126 Turin, Italy; Neuroscience Institute Cavalieri Ottolenghi, University of Turin, 10043 Orbassano (TO), Italy
| | - Yifan Liu
- Department of Molecular Biotechnology and Health Sciences, University of Turin, 10126 Turin, Italy
| | - Fiammetta Vernì
- Department of Biology and Biotechnology "Charles Darwin," Sapienza University, 00185 Rome, Italy
| | - Chiara Merigliano
- Department of Biology and Biotechnology "Charles Darwin," Sapienza University, 00185 Rome, Italy
| | - Silvia Bonaccorsi
- Department of Biology and Biotechnology "Charles Darwin," Sapienza University, 00185 Rome, Italy
| | - Nadia El-Assawy
- Department of Neurology and Neurorehabilitation, San Giuseppe Hospital, Istituto Auxologico Italiano IRCCS, 28824 Piancavallo (VB), Italy
| | - Lorenzo Priano
- Department of Neurology and Neurorehabilitation, San Giuseppe Hospital, Istituto Auxologico Italiano IRCCS, 28824 Piancavallo (VB), Italy; Department of Neuroscience, University of Torino, 10126 Torino, Italy
| | - Marta Gai
- Department of Molecular Biotechnology and Health Sciences, University of Turin, 10126 Turin, Italy
| | - Gaia Elena Berto
- Department of Molecular Biotechnology and Health Sciences, University of Turin, 10126 Turin, Italy; Neuroscience Institute Cavalieri Ottolenghi, University of Turin, 10043 Orbassano (TO), Italy
| | - Alessandra Maria Adelaide Chiotto
- Department of Molecular Biotechnology and Health Sciences, University of Turin, 10126 Turin, Italy; Neuroscience Institute Cavalieri Ottolenghi, University of Turin, 10043 Orbassano (TO), Italy
| | - Francesco Sgrò
- Department of Molecular Biotechnology and Health Sciences, University of Turin, 10126 Turin, Italy
| | - Alessia Caramello
- Department of Molecular Biotechnology and Health Sciences, University of Turin, 10126 Turin, Italy
| | - Laura Tasca
- Department of Molecular Biotechnology and Health Sciences, University of Turin, 10126 Turin, Italy; Neuroscience Institute Cavalieri Ottolenghi, University of Turin, 10043 Orbassano (TO), Italy
| | - Ugo Ala
- Department of Molecular Biotechnology and Health Sciences, University of Turin, 10126 Turin, Italy
| | - Francesco Neri
- Human Genetics Foundation (HuGeF), via Nizza 52, 10126 Torino, Italy
| | | | - Alessandro Mauro
- Department of Neurology and Neurorehabilitation, San Giuseppe Hospital, Istituto Auxologico Italiano IRCCS, 28824 Piancavallo (VB), Italy; Department of Neuroscience, University of Torino, 10126 Torino, Italy
| | - Stephan Geley
- Division of Molecular Pathophysiology, Biocenter, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Maurizio Gatti
- Department of Biology and Biotechnology "Charles Darwin," Sapienza University, 00185 Rome, Italy; Institute of Molecular Biology and Pathology (IBPM), CNR, 00185 Rome, Italy
| | - Ferdinando Di Cunto
- Department of Molecular Biotechnology and Health Sciences, University of Turin, 10126 Turin, Italy; Neuroscience Institute Cavalieri Ottolenghi, University of Turin, 10043 Orbassano (TO), Italy.
| |
Collapse
|
26
|
Duerinckx S, Abramowicz M. The genetics of congenitally small brains. Semin Cell Dev Biol 2017; 76:76-85. [PMID: 28912110 DOI: 10.1016/j.semcdb.2017.09.015] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Revised: 09/05/2017] [Accepted: 09/08/2017] [Indexed: 12/14/2022]
Abstract
Primary microcephaly (PM) refers to a congenitally small brain, resulting from insufficient prenatal production of neurons, and serves as a model disease for brain volumic development. Known PM genes delineate several cellular pathways, among which the centriole duplication pathway, which provide interesting clues about the cellular mechanisms involved. The general interest of the genetic dissection of PM is illustrated by the convergence of Zika virus infection and PM gene mutations on congenital microcephaly, with CENPJ/CPAP emerging as a key target. Physical (protein-protein) and genetic (digenic inheritance) interactions of Wdr62 and Aspm have been demonstrated in mice, and should now be sought in humans using high throughput parallel sequencing of multiple PM genes in PM patients and control subjects, in order to categorize mutually interacting genes, hence delineating functional pathways in vivo in humans.
Collapse
Affiliation(s)
- Sarah Duerinckx
- IRIBHM, Université Libre de Bruxelles, Route de Lennik 808, 1070 Brussels, Belgium.
| | - Marc Abramowicz
- IRIBHM, Université Libre de Bruxelles, Route de Lennik 808, 1070 Brussels, Belgium; Department of Medical Genetics, Hôpital Erasme, Université Libre de Bruxelles, Route de Lennik 808, 1070 Brussels, Belgium.
| |
Collapse
|
27
|
Breuss MW, Leca I, Gstrein T, Hansen AH, Keays DA. Tubulins and brain development - The origins of functional specification. Mol Cell Neurosci 2017; 84:58-67. [PMID: 28347630 DOI: 10.1016/j.mcn.2017.03.002] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2016] [Revised: 02/28/2017] [Accepted: 03/05/2017] [Indexed: 10/19/2022] Open
Abstract
The development of the vertebrate central nervous system is reliant on a complex cascade of biological processes that include mitotic division, relocation of migrating neurons, and the extension of dendritic and axonal processes. Each of these cellular events requires the diverse functional repertoire of the microtubule cytoskeleton for the generation of forces, assembly of macromolecular complexes and transport of molecules and organelles. The tubulins are a multi-gene family that encode for the constituents of microtubules, and have been implicated in a spectrum of neurological disorders. Evidence is building that different tubulins tune the functional properties of the microtubule cytoskeleton dependent on the cell type, developmental profile and subcellular localisation. Here we review of the origins of the functional specification of the tubulin gene family in the developing brain at a transcriptional, translational, and post-transcriptional level. We remind the reader that tubulins are not just loading controls for your average Western blot.
Collapse
Affiliation(s)
- Martin W Breuss
- Department of Neurosciences, University of California, San Diego, La Jolla, CA 92093, USA
| | - Ines Leca
- Research Institute of Molecular Pathology, Vienna Biocenter (VBC), Dr Bohr-Gasse 7, Vienna 1030, Austria
| | - Thomas Gstrein
- Research Institute of Molecular Pathology, Vienna Biocenter (VBC), Dr Bohr-Gasse 7, Vienna 1030, Austria
| | - Andi H Hansen
- Research Institute of Molecular Pathology, Vienna Biocenter (VBC), Dr Bohr-Gasse 7, Vienna 1030, Austria; Institute of Science and Technology Austria, Klosterneuburg 3400, Austria
| | - David A Keays
- Research Institute of Molecular Pathology, Vienna Biocenter (VBC), Dr Bohr-Gasse 7, Vienna 1030, Austria.
| |
Collapse
|
28
|
Abstract
ABSTRACT
Cell division controls the faithful segregation of genomic and cytoplasmic materials between the two nascent daughter cells. Members of the Aurora, Polo and cyclin-dependent (Cdk) kinase families are known to regulate multiple events throughout cell division, whereas another kinase, citron kinase (CIT-K), for a long time has been considered to function solely during cytokinesis, the last phase of cell division. CIT-K was originally proposed to regulate the ingression of the cleavage furrow that forms at the equatorial cortex of the dividing cell after chromosome segregation. However, studies in the last decade have clarified that this kinase is, instead, required for the organization of the midbody in late cytokinesis, and also revealed novel functions of CIT-K earlier in mitosis and in DNA damage control. Moreover, CIT-K mutations have recently been linked to the development of human microcephaly, and CIT-K has been identified as a potential target in cancer therapy. In this Commentary, I describe and re-evaluate the functions and regulation of CIT-K during cell division and its involvement in human disease. Finally, I offer my perspectives on the open questions and future challenges that are necessary to address, in order to fully understand this important and yet unjustly neglected mitotic kinase.
Collapse
Affiliation(s)
- Pier Paolo D'Avino
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge, CB2 1QP, UK
| |
Collapse
|
29
|
McKenzie C, Bassi ZI, Debski J, Gottardo M, Callaini G, Dadlez M, D'Avino PP. Cross-regulation between Aurora B and Citron kinase controls midbody architecture in cytokinesis. Open Biol 2016; 6:rsob.160019. [PMID: 27009191 PMCID: PMC4821246 DOI: 10.1098/rsob.160019] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Cytokinesis culminates in the final separation, or abscission, of the two daughter cells at the end of cell division. Abscission relies on an organelle, the midbody, which forms at the intercellular bridge and is composed of various proteins arranged in a precise stereotypic pattern. The molecular mechanisms controlling midbody organization and function, however, are obscure. Here we show that proper midbody architecture requires cross-regulation between two cell division kinases, Citron kinase (CIT-K) and Aurora B, the kinase component of the chromosomal passenger complex (CPC). CIT-K interacts directly with three CPC components and is required for proper midbody architecture and the orderly arrangement of midbody proteins, including the CPC. In addition, we show that CIT-K promotes Aurora B activity through phosphorylation of the INCENP CPC subunit at the TSS motif. In turn, Aurora B controls CIT-K localization and association with its central spindle partners through phosphorylation of CIT-K's coiled coil domain. Our results identify, for the first time, a cross-regulatory mechanism between two kinases during cytokinesis, which is crucial for establishing the stereotyped organization of midbody proteins.
Collapse
Affiliation(s)
- Callum McKenzie
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge CB2 1QP, UK
| | - Zuni I Bassi
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge CB2 1QP, UK
| | - Janusz Debski
- Mass Spectrometry Laboratory, Institute of Biochemistry and Biophysics, Warszawa 02-106, Poland
| | - Marco Gottardo
- Department of Life Sciences, University of Siena, Via A. Moro 4, Siena 53100, Italy
| | - Giuliano Callaini
- Department of Life Sciences, University of Siena, Via A. Moro 4, Siena 53100, Italy
| | - Michal Dadlez
- Mass Spectrometry Laboratory, Institute of Biochemistry and Biophysics, Warszawa 02-106, Poland
| | - Pier Paolo D'Avino
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge CB2 1QP, UK
| |
Collapse
|
30
|
Affiliation(s)
- Marta Gai
- a Dept. of Molecular Biotechnology and Health Sciences , University of Turin , Turin , Italy
| | - Ferdinando Di Cunto
- a Dept. of Molecular Biotechnology and Health Sciences , University of Turin , Turin , Italy
| |
Collapse
|
31
|
ZIKA virus elicits P53 activation and genotoxic stress in human neural progenitors similar to mutations involved in severe forms of genetic microcephaly. Cell Death Dis 2016; 7:e2440. [PMID: 27787521 PMCID: PMC5133962 DOI: 10.1038/cddis.2016.266] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Revised: 07/20/2016] [Accepted: 07/26/2016] [Indexed: 12/30/2022]
Abstract
Epidemiological evidence from the current outbreak of Zika virus (ZIKV) and recent studies in animal models indicate a strong causal link between ZIKV and microcephaly. ZIKV infection induces cell-cycle arrest and apoptosis in proliferating neural progenitors. However, the mechanisms leading to these phenotypes are still largely obscure. In this report, we explored the possible similarities between transcriptional responses induced by ZIKV in human neural progenitors and those elicited by three different genetic mutations leading to severe forms of microcephaly in mice. We found that the strongest similarity between all these conditions is the activation of common P53 downstream genes. In agreement with these observations, we report that ZIKV infection increases total P53 levels and nuclear accumulation, as well as P53 Ser15 phosphorylation, correlated with genotoxic stress and apoptosis induction. Interestingly, increased P53 activation and apoptosis are induced not only in cells expressing high levels of viral antigens but also in cells showing low or undetectable levels of the same proteins. These results indicate that P53 activation is an early and specific event in ZIKV-infected cells, which could result from cell-autonomous and/or non-cell-autonomous mechanisms. Moreover, we highlight a small group of P53 effector proteins that could act as critical mediators, not only in ZIKV-induced microcephaly but also in many genetic microcephaly syndromes.
Collapse
|
32
|
Gai M, Bianchi FT, Vagnoni C, Vernì F, Bonaccorsi S, Pasquero S, Berto GE, Sgrò F, Chiotto AM, Annaratone L, Sapino A, Bergo A, Landsberger N, Bond J, Huttner WB, Di Cunto F. ASPM and CITK regulate spindle orientation by affecting the dynamics of astral microtubules. EMBO Rep 2016; 17:1396-1409. [PMID: 27562601 DOI: 10.15252/embr.201541823] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2015] [Accepted: 07/26/2016] [Indexed: 11/09/2022] Open
Abstract
Correct orientation of cell division is considered an important factor for the achievement of normal brain size, as mutations in genes that affect this process are among the leading causes of microcephaly. Abnormal spindle orientation is associated with reduction of the neuronal progenitor symmetric divisions, premature cell cycle exit, and reduced neurogenesis. This mechanism has been involved in microcephaly resulting from mutation of ASPM, the most frequently affected gene in autosomal recessive human primary microcephaly (MCPH), but it is presently unknown how ASPM regulates spindle orientation. In this report, we show that ASPM may control spindle positioning by interacting with citron kinase (CITK), a protein whose loss is also responsible for severe microcephaly in mammals. We show that the absence of CITK leads to abnormal spindle orientation in mammals and insects. In mouse cortical development, this phenotype correlates with increased production of basal progenitors. ASPM is required to recruit CITK at the spindle, and CITK overexpression rescues ASPM phenotype. ASPM and CITK affect the organization of astral microtubules (MT), and low doses of MT-stabilizing drug revert the spindle orientation phenotype produced by their knockdown. Finally, CITK regulates both astral-MT nucleation and stability. Our results provide a functional link between two established microcephaly proteins.
Collapse
Affiliation(s)
- Marta Gai
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Federico T Bianchi
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Cristiana Vagnoni
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Fiammetta Vernì
- Department of Biology and Biotechnologies "C. Darwin", Sapienza, Università di Roma, Rome, Italy
| | - Silvia Bonaccorsi
- Department of Biology and Biotechnologies "C. Darwin", Sapienza, Università di Roma, Rome, Italy
| | - Selina Pasquero
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Gaia E Berto
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Francesco Sgrò
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Alessandra Ma Chiotto
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Laura Annaratone
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Anna Sapino
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Anna Bergo
- San Raffaele Rett Research Unit, Division of Neuroscience, San Raffaele Scientific Institute, Milan, Italy
| | - Nicoletta Landsberger
- San Raffaele Rett Research Unit, Division of Neuroscience, San Raffaele Scientific Institute, Milan, Italy
| | - Jacqueline Bond
- Leeds Institute of Biomedical and Clinical Sciences, University of Leeds, Leeds, UK
| | - Wieland B Huttner
- Max-Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Ferdinando Di Cunto
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| |
Collapse
|
33
|
Jungas T, Perchey RT, Fawal M, Callot C, Froment C, Burlet-Schiltz O, Besson A, Davy A. Eph-mediated tyrosine phosphorylation of citron kinase controls abscission. J Cell Biol 2016; 214:555-69. [PMID: 27551053 PMCID: PMC5004443 DOI: 10.1083/jcb.201602057] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Accepted: 07/25/2016] [Indexed: 12/30/2022] Open
Abstract
Abscission is the last step of cytokinesis, allowing the physical separation of daughter cells at the end of cell division. It has been considered a cell autonomous process, yet Jungas et al. report that Ephrin/Eph signaling controls the completion of abscission. Cytokinesis is the last step of cell division, culminating in the physical separation of daughter cells at the end of mitosis. Cytokinesis is a tightly regulated process that until recently was mostly viewed as a cell-autonomous event. Here, we investigated the role of Ephrin/Eph signaling, a well-known local cell-to-cell communication pathway, in cell division. We show that activation of Eph signaling in vitro leads to multinucleation and polyploidy, and we demonstrate that this is caused by alteration of the ultimate step of cytokinesis, abscission. Control of abscission requires Eph kinase activity, and Src and citron kinase (CitK) are downstream effectors in the Eph-induced signal transduction cascade. CitK is phosphorylated on tyrosines in neural progenitors in vivo, and Src kinase directly phosphorylates CitK. We have identified the specific tyrosine residues of CitK that are phosphorylated and show that tyrosine phosphorylation of CitK impairs cytokinesis. Finally, we show that, similar to CitK, Ephrin/Eph signaling controls neuronal ploidy in the developing neocortex. Our study indicates that CitK integrates intracellular and extracellular signals provided by the local environment to coordinate completion of cytokinesis.
Collapse
Affiliation(s)
- Thomas Jungas
- Centre de Biologie du Développement, Centre de Biologie Intégrative, Université de Toulouse, Centre National de la Recherche Scientifique, Université Paul Sabatier, 31062 Toulouse, France
| | - Renaud T Perchey
- Institut National de la Santé et de la Recherche Médicale Unité Mixte de Recherche 1037, Cancer Research Center of Toulouse, 31037 Toulouse, France Centre National de la Recherche Scientifique, ERL 5294, Université de Toulouse, Université Paul Sabatier, 31037 Toulouse, France
| | - Mohamad Fawal
- Centre de Biologie du Développement, Centre de Biologie Intégrative, Université de Toulouse, Centre National de la Recherche Scientifique, Université Paul Sabatier, 31062 Toulouse, France
| | - Caroline Callot
- Institut National de la Santé et de la Recherche Médicale Unité Mixte de Recherche 1037, Cancer Research Center of Toulouse, 31037 Toulouse, France Centre National de la Recherche Scientifique, ERL 5294, Université de Toulouse, Université Paul Sabatier, 31037 Toulouse, France
| | - Carine Froment
- Centre National de la Recherche Scientifique, Institut de Pharmacologie et de Biologie Structurale, 31077 Toulouse, France
| | - Odile Burlet-Schiltz
- Centre National de la Recherche Scientifique, Institut de Pharmacologie et de Biologie Structurale, 31077 Toulouse, France
| | - Arnaud Besson
- Institut National de la Santé et de la Recherche Médicale Unité Mixte de Recherche 1037, Cancer Research Center of Toulouse, 31037 Toulouse, France Centre National de la Recherche Scientifique, ERL 5294, Université de Toulouse, Université Paul Sabatier, 31037 Toulouse, France
| | - Alice Davy
- Centre de Biologie du Développement, Centre de Biologie Intégrative, Université de Toulouse, Centre National de la Recherche Scientifique, Université Paul Sabatier, 31062 Toulouse, France
| |
Collapse
|
34
|
Basit S, Al-Harbi KM, Alhijji SAM, Albalawi AM, Alharby E, Eldardear A, Samman MI. CIT, a gene involved in neurogenic cytokinesis, is mutated in human primary microcephaly. Hum Genet 2016; 135:1199-207. [DOI: 10.1007/s00439-016-1724-0] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2016] [Accepted: 08/09/2016] [Indexed: 12/22/2022]
|
35
|
Li H, Bielas S, Zaki M, Ismail S, Farfara D, Um K, Rosti R, Scott E, Tu S, Chi N, Gabriel S, Erson-Omay E, Ercan-Sencicek A, Yasuno K, Çağlayan A, Kaymakçalan H, Ekici B, Bilguvar K, Gunel M, Gleeson J. Biallelic Mutations in Citron Kinase Link Mitotic Cytokinesis to Human Primary Microcephaly. Am J Hum Genet 2016; 99:501-10. [PMID: 27453578 DOI: 10.1016/j.ajhg.2016.07.004] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2016] [Accepted: 07/05/2016] [Indexed: 10/21/2022] Open
Abstract
Cell division terminates with cytokinesis and cellular separation. Autosomal-recessive primary microcephaly (MCPH) is a neurodevelopmental disorder characterized by a reduction in brain and head size at birth in addition to non-progressive intellectual disability. MCPH is genetically heterogeneous, and 16 loci are known to be associated with loss-of-function mutations predominantly affecting centrosomal-associated proteins, but the multiple roles of centrosomes in cellular function has left questions about etiology. Here, we identified three families affected by homozygous missense mutations in CIT, encoding citron rho-interacting kinase (CIT), which has established roles in cytokinesis. All mutations caused substitution of conserved amino acid residues in the kinase domain and impaired kinase activity. Neural progenitors that were differentiated from induced pluripotent stem cells (iPSCs) derived from individuals with these mutations exhibited abnormal cytokinesis with delayed mitosis, multipolar spindles, and increased apoptosis, rescued by CRISPR/Cas9 genome editing. Our results highlight the importance of cytokinesis in the pathology of primary microcephaly.
Collapse
|