1
|
Pallavicini G, Moccia A, Iegiani G, Parolisi R, Peirent ER, Berto GE, Lorenzati M, Tshuva RY, Ferraro A, Balzac F, Turco E, Salvi SU, Myklebust HF, Wang S, Eisenberg J, Chitale M, Girgla NS, Boda E, Reiner O, Buffo A, Di Cunto F, Bielas SL. Modeling primary microcephaly with human brain organoids reveals fundamental roles of CIT kinase activity. J Clin Invest 2024; 134:e175435. [PMID: 39316437 PMCID: PMC11527453 DOI: 10.1172/jci175435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 09/06/2024] [Indexed: 09/26/2024] Open
Abstract
Brain size and cellular heterogeneity are tightly regulated by species-specific proliferation and differentiation of multipotent neural progenitor cells (NPCs). Errors in this process are among the mechanisms of primary hereditary microcephaly (MCPH), a group of disorders characterized by reduced brain size and intellectual disability. Biallelic citron rho-interacting serine/threonine kinase (CIT) missense variants that disrupt kinase function (CITKI/KI) and frameshift loss-of-function variants (CITFS/FS) are the genetic basis for MCPH17; however, the function of CIT catalytic activity in brain development and NPC cytokinesis is unknown. Therefore, we created the CitKI/KI mouse model and found that it did not phenocopy human microcephaly, unlike biallelic CitFS/FS animals. Nevertheless, both Cit models exhibited binucleation, DNA damage, and apoptosis. To investigate human-specific mechanisms of CIT microcephaly, we generated CITKI/KI and CITFS/FS human forebrain organoids. We found that CITKI/KI and CITFS/FS organoids lost cytoarchitectural complexity, transitioning from pseudostratified to simple neuroepithelium. This change was associated with defects that disrupted the polarity of NPC cytokinesis, in addition to elevating apoptosis. Together, our results indicate that both CIT catalytic and scaffolding functions in NPC cytokinesis are critical for human corticogenesis. Species differences in corticogenesis and the dynamic 3D features of NPC mitosis underscore the utility of human forebrain organoid models for understanding human microcephaly.
Collapse
Affiliation(s)
- Gianmarco Pallavicini
- Neuroscience Institute Cavalieri Ottolenghi, Turin, Italy
- Department of Neuroscience “Rita Levi Montalcini,” University of Turin, Turin, Italy
| | | | - Giorgia Iegiani
- Neuroscience Institute Cavalieri Ottolenghi, Turin, Italy
- Department of Neuroscience “Rita Levi Montalcini,” University of Turin, Turin, Italy
| | - Roberta Parolisi
- Neuroscience Institute Cavalieri Ottolenghi, Turin, Italy
- Department of Neuroscience “Rita Levi Montalcini,” University of Turin, Turin, Italy
| | - Emily R. Peirent
- Neuroscience Graduate Program, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Gaia Elena Berto
- Neuroscience Institute Cavalieri Ottolenghi, Turin, Italy
- Department of Neuroscience “Rita Levi Montalcini,” University of Turin, Turin, Italy
| | - Martina Lorenzati
- Neuroscience Institute Cavalieri Ottolenghi, Turin, Italy
- Department of Neuroscience “Rita Levi Montalcini,” University of Turin, Turin, Italy
| | - Rami Y. Tshuva
- Departments of Molecular Genetics and Molecular Neuroscience, Weizmann Institute of Science, Rehovot, Israel
| | - Alessia Ferraro
- Neuroscience Institute Cavalieri Ottolenghi, Turin, Italy
- Department of Neuroscience “Rita Levi Montalcini,” University of Turin, Turin, Italy
| | - Fiorella Balzac
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Emilia Turco
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | | | | | | | - Julia Eisenberg
- Department of Human Genetics and
- Department of Pediatrics, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | | | | | - Enrica Boda
- Neuroscience Institute Cavalieri Ottolenghi, Turin, Italy
- Department of Neuroscience “Rita Levi Montalcini,” University of Turin, Turin, Italy
| | - Orly Reiner
- Departments of Molecular Genetics and Molecular Neuroscience, Weizmann Institute of Science, Rehovot, Israel
| | - Annalisa Buffo
- Neuroscience Institute Cavalieri Ottolenghi, Turin, Italy
- Department of Neuroscience “Rita Levi Montalcini,” University of Turin, Turin, Italy
| | - Ferdinando Di Cunto
- Neuroscience Institute Cavalieri Ottolenghi, Turin, Italy
- Department of Neuroscience “Rita Levi Montalcini,” University of Turin, Turin, Italy
| | - Stephanie L. Bielas
- Department of Human Genetics and
- Neuroscience Graduate Program, University of Michigan Medical School, Ann Arbor, Michigan, USA
- Department of Pediatrics, University of Michigan Medical School, Ann Arbor, Michigan, USA
| |
Collapse
|
2
|
Maw JJ, Coker JA, Arya T, Goins CM, Sonawane D, Han SH, Rees MG, Ronan MM, Roth JA, Wang NS, Heemers HV, Macdonald JD, Stauffer SR. Discovery and Characterization of Selective, First-in-Class Inhibitors of Citron Kinase. J Med Chem 2024; 67:2631-2666. [PMID: 38330278 DOI: 10.1021/acs.jmedchem.3c01807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2024]
Abstract
Citron kinase (CITK) is an AGC-family serine/threonine kinase that regulates cytokinesis. Despite knockdown experiments implicating CITK as an anticancer target, no selective CITK inhibitors exist. We transformed a previously reported kinase inhibitor with weak off-target CITK activity into a first-in-class CITK chemical probe, C3TD879. C3TD879 is a Type I kinase inhibitor which potently inhibits CITK catalytic activity (biochemical IC50 = 12 nM), binds directly to full-length human CITK in cells (NanoBRET Kd < 10 nM), and demonstrates favorable DMPK properties for in vivo evaluation. We engineered exquisite selectivity for CITK (>17-fold versus 373 other human kinases), making C3TD879 the first chemical probe suitable for interrogating the complex biology of CITK. Our small-molecule CITK inhibitors could not phenocopy the effects of CITK knockdown in cell proliferation, cell cycle progression, or cytokinesis assays, providing preliminary evidence that the structural roles of CITK may be more important than its kinase activity.
Collapse
Affiliation(s)
- Joshua J Maw
- Center for Therapeutics Discovery, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195, United States
| | - Jesse A Coker
- Center for Therapeutics Discovery, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195, United States
| | - Tarun Arya
- Center for Therapeutics Discovery, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195, United States
| | - Christopher M Goins
- Center for Therapeutics Discovery, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195, United States
| | - Dhiraj Sonawane
- Center for Therapeutics Discovery, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195, United States
| | - Sang Hoon Han
- Center for Therapeutics Discovery, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195, United States
| | - Matthew G Rees
- Broad Institute of MIT and Harvard, 415 Main Street, Cambridge Massachusetts 02142, United States
| | - Melissa M Ronan
- Broad Institute of MIT and Harvard, 415 Main Street, Cambridge Massachusetts 02142, United States
| | - Jennifer A Roth
- Broad Institute of MIT and Harvard, 415 Main Street, Cambridge Massachusetts 02142, United States
| | - Nancy S Wang
- Center for Therapeutics Discovery, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195, United States
| | - Hannelore V Heemers
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195, United States
| | - Jonathan D Macdonald
- Center for Therapeutics Discovery, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195, United States
| | - Shaun R Stauffer
- Center for Therapeutics Discovery, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195, United States
| |
Collapse
|
3
|
Liu L, Tang H, Wang K, Liu J, Luo N, Jin G. A three-gene signature reveals changes in the tumor immune microenvironment in the progression from NAFLD to HCC. Sci Rep 2023; 13:22295. [PMID: 38102321 PMCID: PMC10724126 DOI: 10.1038/s41598-023-49358-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 12/07/2023] [Indexed: 12/17/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the most dangerous malignant tumors. The incidence rates of obesity related NAFLD and NASH are increasing year by year, and they are the main risk factors for HCC at present. Finding the mechanism of malignant transformation of NAFLD and NASH is helpful for early prevention and diagnosis. In this study, we performed differential analysis using NAFLD data, NASH data, and HCC data to identify crossover differential genes. Then, using the clinical data of TCGA, a prognostic risk prediction model of three genes (TEAD4, SOCS2, CIT) was constructed, and survival analysis and receiver operating characteristic curves were drawn. The prognostic model was validated using ICGC, GSE116174 and GSE54236 datasets. In addition, we assessed immune status and function in high- and low-risk populations using a prognostic model. Moreover, we assessed the expression of CIT in clinical samples and HCC cell lines and validated its role in HCC development. Our study elucidates the important role of the tumor immune microenvironment in the development of NAFLD/NASH to HCC, deepens the understanding of the pathogenesis of NAFLD/NASH development to HCC, and is helpful for clinical management and decision-making.
Collapse
Affiliation(s)
- Lijuan Liu
- Guangxi Medical University Cancer Hospital, Nan Ning, Guangxi Zhuang Autonomous Region, China
| | - Haonan Tang
- Guangxi Medical University Cancer Hospital, Nan Ning, Guangxi Zhuang Autonomous Region, China
| | - Kui Wang
- Guangxi Medical University Cancer Hospital, Nan Ning, Guangxi Zhuang Autonomous Region, China
| | - Jiaying Liu
- Guangxi Medical University Cancer Hospital, Nan Ning, Guangxi Zhuang Autonomous Region, China
| | - Ningbin Luo
- Guangxi Medical University Cancer Hospital, Nan Ning, Guangxi Zhuang Autonomous Region, China.
| | - Guanqiao Jin
- Guangxi Medical University Cancer Hospital, Nan Ning, Guangxi Zhuang Autonomous Region, China.
| |
Collapse
|
4
|
Rawat C, Ben-Salem S, Singh N, Chauhan G, Rabljenovic A, Vaghela V, Venkadakrishnan VB, Macdonald JD, Dahiya UR, Ghanem Y, Bachour S, Su Y, DePriest AD, Lee S, Muldong M, Kim HT, Kumari S, Valenzuela MM, Zhang D, Hu Q, Cortes Gomez E, Dehm SM, Zoubeidi A, Jamieson CAM, Nicolas M, McKenney J, Willard B, Klein EA, Magi-Galluzzi C, Stauffer SR, Liu S, Heemers HV. Prostate Cancer Progression Relies on the Mitotic Kinase Citron Kinase. Cancer Res 2023; 83:4142-4160. [PMID: 37801613 PMCID: PMC10841833 DOI: 10.1158/0008-5472.can-23-0883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 08/14/2023] [Accepted: 10/03/2023] [Indexed: 10/08/2023]
Abstract
Prostate cancer remains the second leading cause of cancer death in men in Western cultures. A deeper understanding of the mechanisms by which prostate cancer cells divide to support tumor growth could help devise strategies to overcome treatment resistance and improve survival. Here, we identified that the mitotic AGC family protein kinase citron kinase (CIT) is a pivotal regulator of prostate cancer growth that mediates prostate cancer cell interphase progression. Increased CIT expression correlated with prostate cancer growth induction and aggressive prostate cancer progression, and CIT was overexpressed in prostate cancer compared with benign prostate tissue. CIT overexpression was controlled by an E2F2-Skp2-p27 signaling axis and conferred resistance to androgen-targeted treatment strategies. The effects of CIT relied entirely on its kinase activity. Conversely, CIT silencing inhibited the growth of cell lines and xenografts representing different stages of prostate cancer progression and treatment resistance but did not affect benign epithelial prostate cells or nonprostatic normal cells, indicating a potential therapeutic window for CIT inhibition. CIT kinase activity was identified as druggable and was potently inhibited by the multikinase inhibitor OTS-167, which decreased the proliferation of treatment-resistant prostate cancer cells and patient-derived organoids. Isolation of the in vivo CIT substrates identified proteins involved in diverse cellular functions ranging from proliferation to alternative splicing events that are enriched in treatment-resistant prostate cancer. These findings provide insights into the regulation of aggressive prostate cancer cell behavior by CIT and identify CIT as a functionally diverse and druggable driver of prostate cancer progression. SIGNIFICANCE The poorly characterized protein kinase citron kinase is a therapeutic target in prostate cancer that drives tumor growth by regulating diverse substrates, which control several hallmarks of aggressive prostate cancer progression. See related commentary by Mishra et al., p. 4008.
Collapse
Affiliation(s)
- Chitra Rawat
- Department of Cancer Biology, Cleveland Clinic, Cleveland, Ohio
| | - Salma Ben-Salem
- Department of Cancer Biology, Cleveland Clinic, Cleveland, Ohio
| | - Nidhi Singh
- Department of Cancer Biology, Cleveland Clinic, Cleveland, Ohio
| | - Gaurav Chauhan
- Department of Cancer Biology, Cleveland Clinic, Cleveland, Ohio
| | | | - Vishwa Vaghela
- Department of Cancer Biology, Cleveland Clinic, Cleveland, Ohio
| | - Varadha Balaji Venkadakrishnan
- Department of Cancer Biology, Cleveland Clinic, Cleveland, Ohio
- Department of Biological, Geological and Environmental Sciences, Cleveland State University, Cleveland, Ohio
| | | | - Ujjwal R Dahiya
- Department of Cancer Biology, Cleveland Clinic, Cleveland, Ohio
| | - Yara Ghanem
- Department of Cancer Biology, Cleveland Clinic, Cleveland, Ohio
| | - Salam Bachour
- Cleveland Clinic Lerner College of Medicine, Cleveland Clinic, Cleveland, Ohio
| | - Yixue Su
- Department of Cancer Biology, Cleveland Clinic, Cleveland, Ohio
| | - Adam D DePriest
- Department of Cancer Genetics, Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | - Sanghee Lee
- Department of Urology, UC San Diego, La Jolla, California
| | | | - Hyun-Tae Kim
- Department of Urology, UC San Diego, La Jolla, California
- Department of Urology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Sangeeta Kumari
- Department of Cancer Biology, Cleveland Clinic, Cleveland, Ohio
| | | | - Dingxiao Zhang
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, New York
- School of Biomedical Sciences, Hunan University, Changsa, China
| | - Qiang Hu
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | - Eduardo Cortes Gomez
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | - Scott M Dehm
- Masonic Cancer Center and Departments of Laboratory Medicine and Pathology and Urology, University of Minnesota, Minneapolis, Minnesota
| | - Amina Zoubeidi
- Vancouver Prostate Centre and Department of Urologic Sciences, University of British Columbia, Canada
| | | | - Marlo Nicolas
- Department of Anatomic Pathology, Cleveland Clinic, Cleveland, Ohio
| | - Jesse McKenney
- Department of Anatomic Pathology, Cleveland Clinic, Cleveland, Ohio
| | | | - Eric A Klein
- Department of Urology, Cleveland Clinic, Cleveland, Ohio
| | | | - Shaun R Stauffer
- Center for Therapeutics Discovery, Cleveland Clinic, Cleveland, Ohio
| | - Song Liu
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | | |
Collapse
|
5
|
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
|
6
|
Pallavicini G, Iegiani G, Parolisi R, Ferraro A, Garello F, Bitonto V, Terreno E, Gai M, Di Cunto F. Lestaurtinib inhibits Citron kinase activity and medulloblastoma growth through induction of DNA damage, apoptosis and cytokinesis failure. Front Oncol 2023; 13:1202585. [PMID: 37404750 PMCID: PMC10315473 DOI: 10.3389/fonc.2023.1202585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Accepted: 05/30/2023] [Indexed: 07/06/2023] Open
Abstract
Introduction Medulloblastoma (MB), the most common malignant pediatric brain tumor, is currently treated with surgery followed by radiation and chemotherapy, which is accompanied by severe side effects, raising the need for innovative therapies. Disruption of the microcephaly-related gene Citron kinase (CITK) impairs the expansion of xenograft models as well as spontaneous MB arising in transgenic mice. No specific CITK inhibitors are available. Methods Lestaurtinib, a Staurosporine derivative also known as CEP-701, inhibits CITK with IC50 of 90 nM. We therefore tested the biological effects of this molecule on different MB cell lines, as well as in vivo, injecting the drug in MBs arising in SmoA1 transgenic mice. Results Similar to CITK knockdown, treatment of MB cells with 100 nM Lestaurtinib reduces phospho-INCENP levels at the midbody and leads to late cytokinesis failure. Moreover, Lestaurtinib impairs cell proliferation through CITK-sensitive mechanisms. These phenotypes are accompanied by accumulation of DNA double strand breaks, cell cycle block and TP53 superfamily activation in vitro and in vivo. Lestaurtinib treatment reduces tumor growth and increases mice survival. Discussion Our data indicate that Lestaurtinib produces in MB cells poly-pharmacological effects extending beyond the inhibition of its validated targets, supporting the possibility of repositioning this drug for MB treatment.
Collapse
Affiliation(s)
- Gianmarco Pallavicini
- Neuroscience Institute Cavalieri Ottolenghi, Turin, Italy
- Department of Neuroscience ‘Rita Levi Montalcini’, University of Turin, Turin, Italy
| | - Giorgia Iegiani
- Neuroscience Institute Cavalieri Ottolenghi, Turin, Italy
- Department of Neuroscience ‘Rita Levi Montalcini’, University of Turin, Turin, Italy
| | - Roberta Parolisi
- Neuroscience Institute Cavalieri Ottolenghi, Turin, Italy
- Department of Neuroscience ‘Rita Levi Montalcini’, University of Turin, Turin, Italy
| | - Alessia Ferraro
- Neuroscience Institute Cavalieri Ottolenghi, Turin, Italy
- Department of Neuroscience ‘Rita Levi Montalcini’, University of Turin, Turin, Italy
| | - Francesca Garello
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Valeria Bitonto
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Enzo Terreno
- 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
| | - Ferdinando Di Cunto
- Neuroscience Institute Cavalieri Ottolenghi, Turin, Italy
- Department of Neuroscience ‘Rita Levi Montalcini’, University of Turin, Turin, Italy
| |
Collapse
|
7
|
van Bree NFHN, Wilhelm M. The Tumor Microenvironment of Medulloblastoma: An Intricate Multicellular Network with Therapeutic Potential. Cancers (Basel) 2022; 14:5009. [PMID: 36291792 PMCID: PMC9599673 DOI: 10.3390/cancers14205009] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 10/08/2022] [Accepted: 10/10/2022] [Indexed: 11/25/2022] Open
Abstract
Medulloblastoma (MB) is a heterogeneous disease in which survival is highly affected by the underlying subgroup-specific characteristics. Although the current treatment modalities have increased the overall survival rates of MB up to 70-80%, MB remains a major cause of cancer-related mortality among children. This indicates that novel therapeutic approaches against MB are needed. New promising treatment options comprise the targeting of cells and components of the tumor microenvironment (TME). The TME of MB consists of an intricate multicellular network of tumor cells, progenitor cells, astrocytes, neurons, supporting stromal cells, microglia, immune cells, extracellular matrix components, and vasculature systems. In this review, we will discuss all the different components of the MB TME and their role in MB initiation, progression, metastasis, and relapse. Additionally, we briefly introduce the effect that age plays on the TME of brain malignancies and discuss the MB subgroup-specific differences in TME components and how all of these variations could affect the progression of MB. Finally, we highlight the TME-directed treatments, in which we will focus on therapies that are being evaluated in clinical trials.
Collapse
Affiliation(s)
| | - Margareta Wilhelm
- Department of Microbiology, Tumor and Cell Biology (MTC), Karolinska Institute, 17165 Stockholm, Sweden
| |
Collapse
|
8
|
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
|
9
|
Chen J, Li Z, Jia X, Song W, Wu H, Zhu H, Xuan Z, Du Y, Zhu X, Song G, Dong H, Bian S, Wang S, Zhao Y, Xie H, Zheng S, Song P. Targeting anillin inhibits tumorigenesis and tumor growth in hepatocellular carcinoma via impairing cytokinesis fidelity. Oncogene 2022; 41:3118-3130. [PMID: 35477750 DOI: 10.1038/s41388-022-02274-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Revised: 02/21/2022] [Accepted: 03/08/2022] [Indexed: 11/09/2022]
Abstract
Targeting cytokinesis can suppress tumor growth by blocking cell division and promoting apoptosis. We aimed to characterize key cytokinesis regulator in hepatocellular carcinoma (HCC) progression, providing insights into identifying promising HCC therapeutic targets. The unbiased bioinformatic screening identified Anillin actin binding protein (ANLN) as a critical cytokinesis regulator involved in HCC development. Functional assay demonstrated that knockdown of ANLN inhibited HCC growth by inducing cytokinesis failure and DNA damage, leading to multinucleation and mitotic catastrophe. Mechanistically, ANLN acts as a scaffold to strengthen interaction between RACGAP1 and PLK1. ANLN promotes PLK1-mediated RACGAP1 phosphorylation and RhoA activation to ensure cytokinesis fidelity. To explore the function of ANLN in HCC tumorigenesis, we hydrodynamically transfected c-Myc and NRAS plasmids into Anln+/+, Anln+/-, and Anln-/- mice through tail vein injection. Hepatic Anln ablation significantly impaired c-Myc/NRAS-driven hepatocarcinogenesis. Moreover, enhanced hepatic polyploidization was observed in Anln ablation mice, manifesting as increasing proportion of cellular and nuclear polyploidy. Clinically, ANLN is upregulated in human HCC tissues and high level of ANLN is correlated with poor patients' prognosis. Additionally, the proportion of cellular polyploidy decreases during HCC progression and ANLN level is significantly correlated with cellular polyploidy proportion in human HCC samples. In conclusion, ANLN is identified as a key cytokinesis regulator contributing to HCC initiation and progression. Our findings revealed a novel mechanism of ANLN in the regulation of cytokinesis to promote HCC tumorigenesis and growth, suggesting targeting ANLN to inhibit cytokinesis may be a promising therapeutic strategy for HCC.
Collapse
Affiliation(s)
- Jian Chen
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China.,Key Laboratory of the diagnosis and treatment of organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment For Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, China.,Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Hangzhou, Zhejiang Province, China
| | - Zequn Li
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China.,Key Laboratory of the diagnosis and treatment of organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment For Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, China.,Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Hangzhou, Zhejiang Province, China
| | - Xing Jia
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China.,Key Laboratory of the diagnosis and treatment of organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment For Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, China.,Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Hangzhou, Zhejiang Province, China
| | - Wenfeng Song
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China.,Key Laboratory of the diagnosis and treatment of organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment For Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, China.,Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Hangzhou, Zhejiang Province, China
| | - Hao Wu
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China.,Key Laboratory of the diagnosis and treatment of organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment For Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, China.,Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Hangzhou, Zhejiang Province, China
| | - Hai Zhu
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China.,Key Laboratory of the diagnosis and treatment of organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment For Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, China.,Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Hangzhou, Zhejiang Province, China
| | - Zefeng Xuan
- Division of Breast Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yehui Du
- Division of Thyroid Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xingxin Zhu
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China.,Key Laboratory of the diagnosis and treatment of organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment For Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, China.,Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Hangzhou, Zhejiang Province, China
| | - Guangyuan Song
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China.,Key Laboratory of the diagnosis and treatment of organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment For Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, China.,Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Hangzhou, Zhejiang Province, China
| | - Haijiang Dong
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China.,Key Laboratory of the diagnosis and treatment of organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment For Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, China.,Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Hangzhou, Zhejiang Province, China
| | - Suchen Bian
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China.,Key Laboratory of the diagnosis and treatment of organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment For Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, China.,Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Hangzhou, Zhejiang Province, China
| | - Shuo Wang
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yongchao Zhao
- NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China.,Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Haiyang Xie
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China.,Key Laboratory of the diagnosis and treatment of organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment For Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, China.,Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Hangzhou, Zhejiang Province, China
| | - Shusen Zheng
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China. .,NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China. .,Key Laboratory of the diagnosis and treatment of organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment For Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, China. .,Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Hangzhou, Zhejiang Province, China.
| | - Penghong Song
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China. .,NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China. .,Key Laboratory of the diagnosis and treatment of organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment For Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, China. .,Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Hangzhou, Zhejiang Province, China.
| |
Collapse
|
10
|
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
|
11
|
Garrido-Casado M, Asensio-Juárez G, Vicente-Manzanares M. Nonmuscle Myosin II Regulation Directs Its Multiple Roles in Cell Migration and Division. Annu Rev Cell Dev Biol 2021; 37:285-310. [PMID: 34314591 DOI: 10.1146/annurev-cellbio-042721-105528] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Nonmuscle myosin II (NMII) is a multimeric protein complex that generates most mechanical force in eukaryotic cells. NMII function is controlled at three main levels. The first level includes events that trigger conformational changes that extend the complex to enable its assembly into filaments. The second level controls the ATPase activity of the complex and its binding to microfilaments in extended NMII filaments. The third level includes events that modulate the stability and contractility of the filaments. They all work in concert to finely control force generation inside cells. NMII is a common endpoint of mechanochemical signaling pathways that control cellular responses to physical and chemical extracellular cues. Specific phosphorylations modulate NMII activation in a context-dependent manner. A few kinases control these phosphorylations in a spatially, temporally, and lineage-restricted fashion, enabling functional adaptability to the cellular microenvironment. Here, we review mechanisms that control NMII activity in the context of cell migration and division. Expected final online publication date for the Annual Review of Cell and Developmental Biology, Volume 37 is October 2021. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Marina Garrido-Casado
- Molecular Mechanisms Program, Centro de Investigación del Cáncer and Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas, University of Salamanca, 37007 Salamanca, Spain;
| | - Gloria Asensio-Juárez
- Molecular Mechanisms Program, Centro de Investigación del Cáncer and Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas, University of Salamanca, 37007 Salamanca, Spain;
| | - Miguel Vicente-Manzanares
- Molecular Mechanisms Program, Centro de Investigación del Cáncer and Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas, University of Salamanca, 37007 Salamanca, Spain;
| |
Collapse
|
12
|
Llorente-González C, González-Rodríguez M, Vicente-Manzanares M. Targeting cytoskeletal phosphorylation in cancer. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2021; 2:292-308. [PMID: 36046434 PMCID: PMC9400739 DOI: 10.37349/etat.2021.00047] [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: 02/26/2021] [Accepted: 06/01/2021] [Indexed: 11/19/2022] Open
Abstract
Phosphorylation of cytoskeletal proteins regulates the dynamics of polymerization, stability, and disassembly of the different types of cytoskeletal polymers. These control the ability of cells to migrate and divide. Mutations and alterations of the expression levels of multiple protein kinases are hallmarks of most forms of cancer. Thus, altered phosphorylation of cytoskeletal proteins is observed in most cancer cells. These alterations potentially control the ability of cancer cells to divide, invade and form distal metastasis. This review highlights the emergent role of phosphorylation in the control of the function of the different cytoskeletal polymers in cancer cells. It also addresses the potential effect of targeted inhibitors in the normalization of cytoskeletal function.
Collapse
Affiliation(s)
- Clara Llorente-González
- Molecular Mechanisms Program, Centro de Investigación del Cáncer and Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC)-University of Salamanca, 37007 Salamanca, Spain
| | - Marta González-Rodríguez
- Molecular Mechanisms Program, Centro de Investigación del Cáncer and Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC)-University of Salamanca, 37007
| | - Miguel Vicente-Manzanares
- Molecular Mechanisms Program, Centro de Investigación del Cáncer and Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC)-University of Salamanca, 37007 Salamanca, Spain
| |
Collapse
|
13
|
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
|
14
|
Carreno G, Guiho R, Martinez‐Barbera JP. Cell senescence in neuropathology: A focus on neurodegeneration and tumours. Neuropathol Appl Neurobiol 2021; 47:359-378. [PMID: 33378554 PMCID: PMC8603933 DOI: 10.1111/nan.12689] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 12/04/2020] [Accepted: 12/13/2020] [Indexed: 01/10/2023]
Abstract
The study of cell senescence is a burgeoning field. Senescent cells can modify the cellular microenvironment through the secretion of a plethora of biologically active products referred to as the senescence-associated secretory phenotype (SASP). The consequences of these paracrine signals can be either beneficial for tissue homeostasis, if senescent cells are properly cleared and SASP activation is transient, or result in organ dysfunction, when senescent cells accumulate within the tissues and SASP activation is persistent. Several studies have provided evidence for the role of senescence and SASP in promoting age-related diseases or driving organismal ageing. The hype about senescence has been further amplified by the fact that a group of drugs, named senolytics, have been used to successfully ameliorate the burden of age-related diseases and increase health and life span in mice. Ablation of senescent cells in the brain prevents disease progression and improves cognition in murine models of neurodegenerative conditions. The role of senescence in cancer has been more thoroughly investigated, and it is now accepted that senescence is a double-edged sword that can paradoxically prevent or promote tumourigenesis in a context-dependent manner. In addition, senescence induction followed by senolytic treatment is starting to emerge as a novel therapeutic avenue that could improve current anti-cancer therapies and reduce tumour recurrence. In this review, we discuss recent findings supporting the role of cell senescence in the pathogenesis of neurodegenerative diseases and in brain tumours. A better understanding of senescence is likely to result in the development of novel and efficacious anti-senescence therapies against these brain pathologies.
Collapse
Affiliation(s)
- Gabriela Carreno
- Developmental Biology and Cancer ProgrammeBirth Defects Research CentreInstitute of Child Health Great Ormond Street HospitalUniversity College London30 Guilford StreetLondonWC1N 1EHUK
| | - Romain Guiho
- Developmental Biology and Cancer ProgrammeBirth Defects Research CentreInstitute of Child Health Great Ormond Street HospitalUniversity College London30 Guilford StreetLondonWC1N 1EHUK
| | - Juan Pedro Martinez‐Barbera
- Developmental Biology and Cancer ProgrammeBirth Defects Research CentreInstitute of Child Health Great Ormond Street HospitalUniversity College London30 Guilford StreetLondonWC1N 1EHUK
| |
Collapse
|
15
|
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
|
16
|
Davies R, Liu L, Taotao S, Tuano N, Chaturvedi R, Huang KK, Itman C, Mandoli A, Qamra A, Hu C, Powell D, Daly RJ, Tan P, Rosenbluh J. CRISPRi enables isoform-specific loss-of-function screens and identification of gastric cancer-specific isoform dependencies. Genome Biol 2021; 22:47. [PMID: 33499898 PMCID: PMC7836456 DOI: 10.1186/s13059-021-02266-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Accepted: 01/07/2021] [Indexed: 12/26/2022] Open
Abstract
Introduction Genes contain multiple promoters that can drive the expression of various transcript isoforms. Although transcript isoforms from the same gene could have diverse and non-overlapping functions, current loss-of-function methodologies are not able to differentiate between isoform-specific phenotypes. Results Here, we show that CRISPR interference (CRISPRi) can be adopted for targeting specific promoters within a gene, enabling isoform-specific loss-of-function genetic screens. We use this strategy to test functional dependencies of 820 transcript isoforms that are gained in gastric cancer (GC). We identify a subset of GC-gained transcript isoform dependencies, and of these, we validate CIT kinase as a novel GC dependency. We further show that some genes express isoforms with opposite functions. Specifically, we find that the tumour suppressor ZFHX3 expresses an isoform that has a paradoxical oncogenic role that correlates with poor patient outcome. Conclusions Our work finds isoform-specific phenotypes that would not be identified using current loss-of-function approaches that are not designed to target specific transcript isoforms. Supplementary Information The online version contains supplementary material available at 10.1186/s13059-021-02266-6.
Collapse
Affiliation(s)
- Rebecca Davies
- Cancer Research Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, 3800, Australia
| | - Ling Liu
- Cancer Research Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, 3800, Australia
| | - Sheng Taotao
- Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore, 169857, Singapore.,Cancer Science Institute of Singapore, National University of Singapore, Singapore, 117599, Singapore.,Cancer Therapeutics and Stratified Oncology, Genome Institute of Singapore, Singapore, 138672, Singapore.,SingHealth/Duke-NUS Institute of Precision Medicine, National Heart Centre Singapore, Singapore, 169856, Singapore.,Cellular and Molecular Research, National Cancer Centre, Singapore, 169610, Singapore.,Singapore Gastric Cancer Consortium, Singapore, 119074, Singapore
| | - Natasha Tuano
- Cancer Research Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, 3800, Australia
| | - Richa Chaturvedi
- Cancer Research Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, 3800, Australia
| | - Kie Kyon Huang
- Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore, 169857, Singapore.,Cancer Science Institute of Singapore, National University of Singapore, Singapore, 117599, Singapore.,Cancer Therapeutics and Stratified Oncology, Genome Institute of Singapore, Singapore, 138672, Singapore.,SingHealth/Duke-NUS Institute of Precision Medicine, National Heart Centre Singapore, Singapore, 169856, Singapore.,Cellular and Molecular Research, National Cancer Centre, Singapore, 169610, Singapore.,Singapore Gastric Cancer Consortium, Singapore, 119074, Singapore
| | - Catherine Itman
- Functional Genomics Platform, Monash University, Clayton, VIC, 3800, Australia
| | - Amit Mandoli
- Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore, 169857, Singapore
| | - Aditi Qamra
- Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore, 169857, Singapore.,Cancer Science Institute of Singapore, National University of Singapore, Singapore, 117599, Singapore.,Cancer Therapeutics and Stratified Oncology, Genome Institute of Singapore, Singapore, 138672, Singapore.,SingHealth/Duke-NUS Institute of Precision Medicine, National Heart Centre Singapore, Singapore, 169856, Singapore.,Cellular and Molecular Research, National Cancer Centre, Singapore, 169610, Singapore.,Singapore Gastric Cancer Consortium, Singapore, 119074, Singapore
| | - Changyuan Hu
- Cancer Research Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, 3800, Australia
| | - David Powell
- Monash Bioinformatics Platform, Monash University, Clayton, VIC, 3800, Australia
| | - Roger J Daly
- Cancer Research Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, 3800, Australia
| | - Patrick Tan
- Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore, 169857, Singapore. .,Cancer Science Institute of Singapore, National University of Singapore, Singapore, 117599, Singapore. .,Cancer Therapeutics and Stratified Oncology, Genome Institute of Singapore, Singapore, 138672, Singapore. .,SingHealth/Duke-NUS Institute of Precision Medicine, National Heart Centre Singapore, Singapore, 169856, Singapore. .,Cellular and Molecular Research, National Cancer Centre, Singapore, 169610, Singapore. .,Singapore Gastric Cancer Consortium, Singapore, 119074, Singapore.
| | - Joseph Rosenbluh
- Cancer Research Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, 3800, Australia. .,Functional Genomics Platform, Monash University, Clayton, VIC, 3800, Australia.
| |
Collapse
|
17
|
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
|
18
|
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
|
19
|
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
|
20
|
Tamayo-Orrego L, Charron F. Recent advances in SHH medulloblastoma progression: tumor suppressor mechanisms and the tumor microenvironment. F1000Res 2019; 8. [PMID: 31700613 PMCID: PMC6820827 DOI: 10.12688/f1000research.20013.1] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/23/2019] [Indexed: 12/13/2022] Open
Abstract
Medulloblastoma, the most common of the malignant pediatric brain tumors, is a group of four molecularly and clinically distinct cancers with different cells of origin. One of these medulloblastoma groups displays activation of Sonic hedgehog (SHH) signaling and originates from granule cell precursors of the developing cerebellum. Ongoing basic and clinical research efforts are tailored to discover targeted and safer therapies, which rely on the identification of the basic mechanisms regulating tumor initiation, progression, and metastasis. In SHH medulloblastoma, the mechanisms regulating neural progenitor transformation and progression to advanced tumors have been studied in some detail. The present review discusses recent advances on medulloblastoma progression derived from studies using mouse models of SHH medulloblastoma. We focus on mechanisms that regulate progression from precancerous lesions to medulloblastoma, describing novel roles played by tumor suppressor mechanisms and the tumor microenvironment.
Collapse
Affiliation(s)
- Lukas Tamayo-Orrego
- Montreal Clinical Research Institute (IRCM), Montreal, Quebec, Canada.,Integrated Program in Neuroscience, McGill University, Montreal, Quebec, Canada.,Grupo Neuroaprendizaje, Autonomous University of Manizales, Manizales, Colombia
| | - Frédéric Charron
- Montreal Clinical Research Institute (IRCM), Montreal, Quebec, Canada.,Integrated Program in Neuroscience, McGill University, Montreal, Quebec, Canada.,Department of Anatomy and Cell Biology, Division of Experimental Medicine, McGill University, Quebec, Canada.,Department of Medicine, University of Montreal, Montreal, Quebec, Canada
| |
Collapse
|
21
|
Cabel L, Proudhon C, Romano E, Girard N, Lantz O, Stern MH, Pierga JY, Bidard FC. Clinical potential of circulating tumour DNA in patients receiving anticancer immunotherapy. Nat Rev Clin Oncol 2019; 15:639-650. [PMID: 30050094 DOI: 10.1038/s41571-018-0074-3] [Citation(s) in RCA: 142] [Impact Index Per Article: 28.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Considerable interest surrounds the use of immune-checkpoint inhibitors in patients with solid tumours following the demonstration of the impressive clinical efficacy of anti-programmed cell death protein 1 and anti-programmed cell death 1 ligand 1 antibodies in several tumour types. However, the emergence of unexpected tumour response patterns, such as pseudoprogression or hyperprogression, might complicate the management of patients receiving these agents. Analysis of circulating tumour DNA (ctDNA) has been shown to have prognostic value by enabling the detection of residual proliferating disease in the adjuvant setting and estimation of tumour burden in the metastatic setting, which are key stratification biomarkers for use of immune-checkpoint inhibition (ICI). Furthermore, examinations of ctDNA for genetic predictors of responsiveness to immunotherapy, such as mutations, tumour mutational load, and microsatellite instability provide a noninvasive surrogate for tumour biopsy sampling. Proof-of-concept reports have also demonstrated that quantitative changes in ctDNA levels early in the course of disease are a promising tool for the assessment of responsiveness to ICI that might complement standard imaging approaches. Other applications of this technology are also currently under investigation, such as early detection of resistance to immunotherapy and characterization of mechanisms of resistance. The aim of this Review is to summarize available data on the application of ctDNA in patients receiving immunotherapy and to discuss the most promising future directions.
Collapse
Affiliation(s)
- Luc Cabel
- Department of Medical Oncology, Institut Curie, PSL Research University, Paris, France.,Versailles Saint Quentin en Yvelines University, Paris Saclay University, Saint Cloud, Paris, France.,Circulating Tumor Biomarkers Laboratory, Institut Curie, PSL Research University, Paris, France
| | - Charlotte Proudhon
- Circulating Tumor Biomarkers Laboratory, Institut Curie, PSL Research University, Paris, France
| | - Emanuela Romano
- Department of Medical Oncology, Institut Curie, PSL Research University, Paris, France.,INSERM U932, Institut Curie, PSL Research University, Paris, France
| | - Nicolas Girard
- Department of Medical Oncology, Institut Curie, PSL Research University, Paris, France
| | - Olivier Lantz
- INSERM U932, Institut Curie, PSL Research University, Paris, France
| | - Marc-Henri Stern
- INSERM U830, Institut Curie, PSL Research University, Paris, France
| | - Jean-Yves Pierga
- Department of Medical Oncology, Institut Curie, PSL Research University, Paris, France.,Paris Descartes University, Paris, France
| | - François-Clément Bidard
- Department of Medical Oncology, Institut Curie, PSL Research University, Paris, France. .,Versailles Saint Quentin en Yvelines University, Paris Saclay University, Saint Cloud, Paris, France. .,Circulating Tumor Biomarkers Laboratory, Institut Curie, PSL Research University, Paris, France.
| |
Collapse
|
22
|
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
|
23
|
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
|