1
|
Schütt J, Brinkert K, Plis A, Schenk T, Brioli A. Unraveling the complexity of drug resistance mechanisms to SINE, T cell-engaging therapies and CELMoDs in multiple myeloma: a comprehensive review. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2024; 7:26. [PMID: 39050883 PMCID: PMC11267153 DOI: 10.20517/cdr.2024.39] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Revised: 05/30/2024] [Accepted: 06/20/2024] [Indexed: 07/27/2024]
Abstract
Despite significant advances in the understanding of multiple myeloma (MM) biology and the development of novel treatment strategies in the last two decades, MM is still an incurable disease. Novel drugs with alternative mechanisms of action, such as selective inhibitors of nuclear export (SINE), modulators of the ubiquitin pathway [cereblon E3 ligase modulatory drugs (CELMoDs)], and T cell redirecting (TCR) therapy, have led to significant improvement in patient outcomes. However, resistance still emerges, posing a major problem for the treatment of myeloma patients. This review summarizes current data on treatment with SINE, TCR therapy, and CELMoDs and explores their mechanism of resistance. Understanding these resistance mechanisms is critical for developing strategies to overcome treatment failure and improve therapeutic outcomes.
Collapse
Affiliation(s)
- Jacqueline Schütt
- Clinic for Hematology, Hemostasis, Oncology and Stem cell transplantation, Hannover Medical School, Hannover 30625, Germany
- Authors contributed equally
| | - Kerstin Brinkert
- Clinic for Hematology, Hemostasis, Oncology and Stem cell transplantation, Hannover Medical School, Hannover 30625, Germany
- Authors contributed equally
| | - Andrzej Plis
- Clinic for Internal Medicine C, Hematology and Oncology, Greifswald University Medicine, Greifswald 17489, Germany
| | - Tino Schenk
- Clinic of Internal Medicine 2, Department of Hematology and Medical Oncology, Jena University Hospital, Jena 07741, Germany
- Institute of Molecular Cell Biology, CMB, Jena University Hospital, Jena 07741, Germany
| | - Annamaria Brioli
- Clinic for Hematology, Hemostasis, Oncology and Stem cell transplantation, Hannover Medical School, Hannover 30625, Germany
- Clinic for Internal Medicine C, Hematology and Oncology, Greifswald University Medicine, Greifswald 17489, Germany
| |
Collapse
|
2
|
Lai C, Xu L, Dai S. The nuclear export protein exportin-1 in solid malignant tumours: From biology to clinical trials. Clin Transl Med 2024; 14:e1684. [PMID: 38783482 PMCID: PMC11116501 DOI: 10.1002/ctm2.1684] [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: 12/07/2023] [Revised: 04/15/2024] [Accepted: 04/19/2024] [Indexed: 05/25/2024] Open
Abstract
BACKGROUND Exportin-1 (XPO1), a crucial protein regulating nuclear-cytoplasmic transport, is frequently overexpressed in various cancers, driving tumor progression and drug resistance. This makes XPO1 an attractive therapeutic target. Over the past few decades, the number of available nuclear export-selective inhibitors has been increasing. Only KPT-330 (selinexor) has been successfully used for treating haematological malignancies, and KPT-8602 (eltanexor) has been used for treating haematologic tumours in clinical trials. However, the use of nuclear export-selective inhibitors for the inhibition of XPO1 expression has yet to be thoroughly investigated in clinical studies and therapeutic outcomes for solid tumours. METHODS We collected numerous literatures to explain the efficacy of XPO1 Inhibitors in preclinical and clinical studies of a wide range of solid tumours. RESULTS In this review, we focus on the nuclear export function of XPO1 and results from clinical trials of its inhibitors in solid malignant tumours. We summarized the mechanism of action and therapeutic potential of XPO1 inhibitors, as well as adverse effects and response biomarkers. CONCLUSION XPO1 inhibition has emerged as a promising therapeutic strategy in the fight against cancer, offering a novel approach to targeting tumorigenic processes and overcoming drug resistance. SINE compounds have demonstrated efficacy in a wide range of solid tumours, and ongoing research is focused on optimizing their use, identifying response biomarkers, and developing effective combination therapies. KEY POINTS Exportin-1 (XPO1) plays a critical role in mediating nucleocytoplasmic transport and cell cycle. XPO1 dysfunction promotes tumourigenesis and drug resistance within solid tumours. The therapeutic potential and ongoing researches on XPO1 inhibitors in the treatment of solid tumours. Additional researches are essential to address safety concerns and identify biomarkers for predicting patient response to XPO1 inhibitors.
Collapse
Affiliation(s)
- Chuanxi Lai
- Department of Colorectal SurgerySir Run Run Shaw HospitalSchool of MedicineZhejiang UniversityHangzhouChina
- Key Laboratory of Biotherapy of Zhejiang ProvinceHangzhouChina
| | - Lingna Xu
- Department of Colorectal SurgerySir Run Run Shaw HospitalSchool of MedicineZhejiang UniversityHangzhouChina
- Key Laboratory of Biotherapy of Zhejiang ProvinceHangzhouChina
| | - Sheng Dai
- Department of Colorectal SurgerySir Run Run Shaw HospitalSchool of MedicineZhejiang UniversityHangzhouChina
- Key Laboratory of Biotherapy of Zhejiang ProvinceHangzhouChina
| |
Collapse
|
3
|
Yang Y, Guo L, Chen L, Gong B, Jia D, Sun Q. Nuclear transport proteins: structure, function, and disease relevance. Signal Transduct Target Ther 2023; 8:425. [PMID: 37945593 PMCID: PMC10636164 DOI: 10.1038/s41392-023-01649-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2023] [Revised: 09/13/2023] [Accepted: 09/14/2023] [Indexed: 11/12/2023] Open
Abstract
Proper subcellular localization is crucial for the functioning of biomacromolecules, including proteins and RNAs. Nuclear transport is a fundamental cellular process that regulates the localization of many macromolecules within the nuclear or cytoplasmic compartments. In humans, approximately 60 proteins are involved in nuclear transport, including nucleoporins that form membrane-embedded nuclear pore complexes, karyopherins that transport cargoes through these complexes, and Ran system proteins that ensure directed and rapid transport. Many of these nuclear transport proteins play additional and essential roles in mitosis, biomolecular condensation, and gene transcription. Dysregulation of nuclear transport is linked to major human diseases such as cancer, neurodegenerative diseases, and viral infections. Selinexor (KPT-330), an inhibitor targeting the nuclear export factor XPO1 (also known as CRM1), was approved in 2019 to treat two types of blood cancers, and dozens of clinical trials of are ongoing. This review summarizes approximately three decades of research data in this field but focuses on the structure and function of individual nuclear transport proteins from recent studies, providing a cutting-edge and holistic view on the role of nuclear transport proteins in health and disease. In-depth knowledge of this rapidly evolving field has the potential to bring new insights into fundamental biology, pathogenic mechanisms, and therapeutic approaches.
Collapse
Affiliation(s)
- Yang Yang
- Department of Pulmonary and Critical Care Medicine, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Lu Guo
- Department of Pulmonary and Critical Care Medicine, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Lin Chen
- Department of Pulmonary and Critical Care Medicine, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Bo Gong
- The Key Laboratory for Human Disease Gene Study of Sichuan Province and Department of Laboratory Medicine, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Research Unit for Blindness Prevention of Chinese Academy of Medical Sciences (2019RU026), Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu, China
| | - Da Jia
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Department of Pediatrics, West China Second University Hospital, State Key Laboratory of Biotherapy, Sichuan University, Chengdu, China.
| | - Qingxiang Sun
- Department of Pulmonary and Critical Care Medicine, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China.
- Department of Pathology, State Key Laboratory of Biotherapy and Cancer Centre, West China Hospital, Sichuan University, and Collaborative Innovation Centre of Biotherapy, Chengdu, China.
| |
Collapse
|
4
|
Mghezzi-Habellah M, Prochasson L, Jalinot P, Mocquet V. Viral Subversion of the Chromosome Region Maintenance 1 Export Pathway and Its Consequences for the Cell Host. Viruses 2023; 15:2218. [PMID: 38005895 PMCID: PMC10674744 DOI: 10.3390/v15112218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 10/28/2023] [Accepted: 11/01/2023] [Indexed: 11/26/2023] Open
Abstract
In eukaryotic cells, the spatial distribution between cytoplasm and nucleus is essential for cell homeostasis. This dynamic distribution is selectively regulated by the nuclear pore complex (NPC), which allows the passive or energy-dependent transport of proteins between these two compartments. Viruses possess many strategies to hijack nucleocytoplasmic shuttling for the benefit of their viral replication. Here, we review how viruses interfere with the karyopherin CRM1 that controls the nuclear export of protein cargoes. We analyze the fact that the viral hijacking of CRM1 provokes are-localization of numerous cellular factors in a suitable place for specific steps of viral replication. While CRM1 emerges as a critical partner for viruses, it also takes part in antiviral and inflammatory response regulation. This review also addresses how CRM1 hijacking affects it and the benefits of CRM1 inhibitors as antiviral treatments.
Collapse
Affiliation(s)
| | | | | | - Vincent Mocquet
- Laboratoire de Biologie et Modélisation de la Cellule, Ecole Normale Supérieure-Lyon, Université Claude Bernard Lyon, U1293, UMR5239, 69364 Lyon, France; (M.M.-H.); (L.P.); (P.J.)
| |
Collapse
|
5
|
Damizia M, Altieri L, Costanzo V, Lavia P. Distinct Mitotic Functions of Nucleolar and Spindle-Associated Protein 1 (NuSAP1) Are Controlled by Two Consensus SUMOylation Sites. Cells 2023; 12:2545. [PMID: 37947624 PMCID: PMC10650578 DOI: 10.3390/cells12212545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 10/24/2023] [Accepted: 10/25/2023] [Indexed: 11/12/2023] Open
Abstract
Nucleolar and Spindle-Associated Protein 1 (NuSAP1) is an important mitotic regulator, implicated in control of mitotic microtubule stability and chromosome segregation. NuSAP1 regulates these processes by interacting with several protein partners. Its abundance, activity and interactions are therefore tightly regulated during mitosis. Protein conjugation with SUMO (Small Ubiquitin-like MOdifier peptide) is a reversible post-translational modification that modulates rapid changes in the structure, interaction(s) and localization of proteins. NuSAP1 was previously found to interact with RANBP2, a nucleoporin with SUMO ligase and SUMO-stabilizing activity, but how this interaction affects NuSAP1 activity has remained elusive. Here, we show that NuSAP1 interacts with RANBP2 and forms proximity ligation products with SUMO2/3 peptides in a RANBP2-dependent manner at key mitotic sites. A bioinformatic search identified two putative SUMO consensus sites in NuSAP1, within the DNA-binding and the microtubule-binding domains, respectively. Site-specific mutagenesis, and mitotic phenotyping in cell lines expressing each NuSAP1 mutant version, revealed selective roles of each individual site in control of NuSAP1 localization and in generation of specific mitotic defects and distinct fates in daughter cells. These results identify therefore two new regulatory sites for NuSAP1 functions and implicate RANBP2 in control of NuSAP1 activity.
Collapse
Affiliation(s)
- Michela Damizia
- Institute of Molecular Biology and Pathology (IBPM), CNR National Research Council of Italy, 00185 Rome, Italy; (M.D.); (L.A.); (V.C.)
- Department of Biology and Biotechnology “Charles Darwin”, Sapienza University of Rome, 00185 Rome, Italy
- Department of Cellular, Computational and Integrated Biology (CIBIO), University of Trento, 38123 Trento, Italy
| | - Ludovica Altieri
- Institute of Molecular Biology and Pathology (IBPM), CNR National Research Council of Italy, 00185 Rome, Italy; (M.D.); (L.A.); (V.C.)
- Department of Biology and Biotechnology “Charles Darwin”, Sapienza University of Rome, 00185 Rome, Italy
| | - Vincenzo Costanzo
- Institute of Molecular Biology and Pathology (IBPM), CNR National Research Council of Italy, 00185 Rome, Italy; (M.D.); (L.A.); (V.C.)
| | - Patrizia Lavia
- Institute of Molecular Biology and Pathology (IBPM), CNR National Research Council of Italy, 00185 Rome, Italy; (M.D.); (L.A.); (V.C.)
| |
Collapse
|
6
|
Arroyo-Berdugo Y, Sendino M, Greaves D, Nojszewska N, Idilli O, So CW, Di Silvio L, Quartey-Papafio R, Farzaneh F, Rodriguez JA, Calle Y. High Throughput Fluorescence-Based In Vitro Experimental Platform for the Identification of Effective Therapies to Overcome Tumour Microenvironment-Mediated Drug Resistance in AML. Cancers (Basel) 2023; 15:1988. [PMID: 37046649 PMCID: PMC10093176 DOI: 10.3390/cancers15071988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 03/20/2023] [Indexed: 03/29/2023] Open
Abstract
The interactions between Acute Myeloid Leukaemia (AML) leukemic stem cells and the bone marrow (BM) microenvironment play a critical role during AML progression and resistance to drug treatments. Therefore, the identification of novel therapies requires drug-screening methods using in vitro co-culture models that closely recreate the cytoprotective BM setting. We have developed a new fluorescence-based in vitro co-culture system scalable to high throughput for measuring the concomitant effect of drugs on AML cells and the cytoprotective BM microenvironment. eGFP-expressing AML cells are co-cultured in direct contact with mCherry-expressing BM stromal cells for the accurate assessment of proliferation, viability, and signaling in both cell types. This model identified several efficacious compounds that overcome BM stroma-mediated drug resistance against daunorubicin, including the chromosome region maintenance 1 (CRM1/XPO1) inhibitor KPT-330. In silico analysis of genes co-expressed with CRM1, combined with in vitro experiments using our new methodology, also indicates that the combination of KPT-330 with the AURKA pharmacological inhibitor alisertib circumvents the cytoprotection of AML cells mediated by the BM stroma. This new experimental model and analysis provide a more precise screening method for developing improved therapeutics targeting AML cells within the cytoprotective BM microenvironment.
Collapse
Affiliation(s)
- Yoana Arroyo-Berdugo
- School of Health and Life Sciences, University of Roehampton, London SW15 4JD, UK
| | - Maria Sendino
- Department of Genetics, Physical Anthropology and Animal Physiology, University of the Basque Country (UPV/EHU), 48940 Leioa, Spain
| | - David Greaves
- School of Health and Life Sciences, University of Roehampton, London SW15 4JD, UK
| | - Natalia Nojszewska
- School of Health and Life Sciences, University of Roehampton, London SW15 4JD, UK
| | - Orest Idilli
- School of Health and Life Sciences, University of Roehampton, London SW15 4JD, UK
| | - Chi Wai So
- Department of Haemato-Oncology, King’s College London, London SE5 9NU, UK
| | - Lucy Di Silvio
- Faculty of Dentistry, Oral & Craniofacial Sciences, King’s College London, London SE1 9RT, UK
| | | | - Farzin Farzaneh
- Department of Haemato-Oncology, King’s College London, London SE5 9NU, UK
| | - Jose Antonio Rodriguez
- Department of Genetics, Physical Anthropology and Animal Physiology, University of the Basque Country (UPV/EHU), 48940 Leioa, Spain
| | - Yolanda Calle
- School of Health and Life Sciences, University of Roehampton, London SW15 4JD, UK
| |
Collapse
|
7
|
Liu Y, Azizian NG, Sullivan DK, Li Y. mTOR inhibition attenuates chemosensitivity through the induction of chemotherapy resistant persisters. Nat Commun 2022; 13:7047. [PMID: 36396656 PMCID: PMC9671908 DOI: 10.1038/s41467-022-34890-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 11/10/2022] [Indexed: 11/18/2022] Open
Abstract
Chemotherapy can eradicate a majority of cancer cells. However, a small population of tumor cells often survives drug treatments through genetic and/or non-genetic mechanisms, leading to tumor recurrence. Here we report a reversible chemoresistance phenotype regulated by the mTOR pathway. Through a genome-wide CRISPR knockout library screen in pancreatic cancer cells treated with chemotherapeutic agents, we have identified the mTOR pathway as a prominent determinant of chemosensitivity. Pharmacological suppression of mTOR activity in cancer cells from diverse tissue origins leads to the persistence of a reversibly resistant population, which is otherwise eliminated by chemotherapeutic agents. Conversely, activation of the mTOR pathway increases chemosensitivity in vitro and in vivo and predicts better survival among various human cancers. Persister cells display a senescence phenotype. Inhibition of mTOR does not induce cellular senescence per se, but rather promotes the survival of senescent cells through regulation of autophagy and G2/M cell cycle arrest, as revealed by a small-molecule chemical library screen. Thus, mTOR plays a causal yet paradoxical role in regulating chemotherapeutic response; inhibition of the mTOR pathway, while suppressing tumor expansion, facilitates the development of a reversible drug-tolerant senescence state.
Collapse
Affiliation(s)
- Yuanhui Liu
- grid.63368.380000 0004 0445 0041Center for Immunotherapy Research, Houston Methodist Research Institute, Houston, TX 77030 USA ,grid.5386.8000000041936877XDepartment of Medicine, Weill Cornell Medical College, New York, NY 10065 USA
| | - Nancy G. Azizian
- grid.63368.380000 0004 0445 0041Center for Immunotherapy Research, Houston Methodist Research Institute, Houston, TX 77030 USA ,grid.5386.8000000041936877XDepartment of Medicine, Weill Cornell Medical College, New York, NY 10065 USA
| | - Delaney K. Sullivan
- grid.19006.3e0000 0000 9632 6718UCLA-Caltech Medical Scientist Training Program, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095 USA
| | - Yulin Li
- grid.63368.380000 0004 0445 0041Center for Immunotherapy Research, Houston Methodist Research Institute, Houston, TX 77030 USA ,grid.5386.8000000041936877XDepartment of Medicine, Weill Cornell Medical College, New York, NY 10065 USA
| |
Collapse
|
8
|
Stephens DM, Huang Y, Ruppert AS, Walker JS, Canfield D, Cempre CB, Fu Q, Baker S, Hu B, Shah H, Vadeboncoeur R, Rogers KA, Bhat S, Jaglowski SM, Lockman H, Lapalombella R, Byrd JC, Woyach JA. Selinexor Combined with Ibrutinib Demonstrates Tolerability and Safety in Advanced B-Cell Malignancies: A Phase I Study. Clin Cancer Res 2022; 28:3242-3247. [PMID: 35608822 PMCID: PMC9364840 DOI: 10.1158/1078-0432.ccr-21-3867] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 12/22/2021] [Accepted: 05/18/2022] [Indexed: 02/04/2023]
Abstract
PURPOSE Dual blockade of Bruton's tyrosine kinase with ibrutinib and selinexor has potential to deepen responses for patients with chronic lymphocytic leukemia (CLL) and non-Hodgkin lymphoma (NHL). PATIENTS AND METHODS In this phase I study (clinicaltrials.gov: NCT02303392), adult patients with CLL/NHL, relapsed/refractory to ≥1 prior therapy were enrolled. Patients received weekly oral selinexor and daily oral ibrutinib in 28-day cycles until progression or intolerance. Primary objective was to determine MTD. RESULTS Included patients had CLL (n = 16) or NHL (n = 18; 9 Richter transformation, 6 diffuse large B-cell lymphoma, and 3 mantle cell lymphoma). Median prior therapies were 4 (range = 1-14) and 59% previously received ibrutinib. The established MTD was 40 mg of selinexor (days 1, 8, 15) and 420 mg daily ibrutinib. Common nonhematologic adverse events were fatigue (56%), nausea (53%), anorexia (41%), and diarrhea (41%) and were mostly low grade. Overall response rate was 32%. An additional 47% achieved stable disease (SD), some prolonged (up to 36 months). Median progression-free survival for patients with CLL and NHL was 8.9 [95% confidence interval (CI), 3.9-16.1] and 2.7 (95% CI, 0.7-5.4) months, respectively. For patients with CLL who did not receive prior ibrutinib, only 20% (1/5) progressed. Estimated 2-year overall survival was 73.7% (95% CI, 44.1-89.2) and 27.8% (95% CI, 10.1-48.9) for patients with CLL and NHL, respectively. CONCLUSIONS The selinexor and ibrutinib combination has demonstrated tolerability in patients with relapsed/refractory CLL/NHL. Responses were durable. Notable responses were seen in patients with CLL with minimal prior therapy. Future study of this combination will focus on efforts to deepen remissions in patients with CLL receiving ibrutinib therapy.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Qiang Fu
- Ohio State University, Columbus, OH
| | | | - Boyu Hu
- University of Utah, Salt Lake City, UT
| | | | | | | | | | | | | | | | | | | |
Collapse
|
9
|
Lin KH, Rutter JC, Xie A, Killarney ST, Vaganay C, Benaksas C, Ling F, Sodaro G, Meslin PA, Bassil CF, Fenouille N, Hoj J, Washart R, Ang HX, Cerda-Smith C, Chaintreuil P, Jacquel A, Auberger P, Forget A, Itzykson R, Lu M, Lin J, Pierobon M, Sheng Z, Li X, Chilkoti A, Owzar K, Rizzieri DA, Pardee TS, Benajiba L, Petricoin E, Puissant A, Wood KC. P2RY2-AKT activation is a therapeutically actionable consequence of XPO1 inhibition in acute myeloid leukemia. NATURE CANCER 2022; 3:837-851. [PMID: 35668193 PMCID: PMC9949365 DOI: 10.1038/s43018-022-00394-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2021] [Accepted: 05/04/2022] [Indexed: 12/12/2022]
Abstract
Selinexor is a first-in-class inhibitor of the nuclear exportin XPO1 that was recently approved by the US Food and Drug Administration for the treatment of multiple myeloma and diffuse large B-cell lymphoma. In relapsed/refractory acute myeloid leukemia (AML), selinexor has shown promising activity, suggesting that selinexor-based combination therapies may have clinical potential. Here, motivated by the hypothesis that selinexor's nuclear sequestration of diverse substrates imposes pleiotropic fitness effects on AML cells, we systematically catalog the pro- and anti-fitness consequences of selinexor treatment. We discover that selinexor activates PI3Kγ-dependent AKT signaling in AML by upregulating the purinergic receptor P2RY2. Inhibiting this axis potentiates the anti-leukemic effects of selinexor in AML cell lines, patient-derived primary cultures and multiple mouse models of AML. In a syngeneic, MLL-AF9-driven mouse model of AML, treatment with selinexor and ipatasertib outperforms both standard-of-care chemotherapy and chemotherapy with selinexor. Together, these findings establish drug-induced P2RY2-AKT signaling as an actionable consequence of XPO1 inhibition in AML.
Collapse
Affiliation(s)
- Kevin H Lin
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC, USA
| | - Justine C Rutter
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC, USA
| | - Abigail Xie
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC, USA
| | - Shane T Killarney
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC, USA
| | - Camille Vaganay
- Université de Paris, Génomes, Biologie Cellulaire et Thérapeutique U944, INSERM, CNRS, Paris, France
| | - Chaima Benaksas
- Université de Paris, Génomes, Biologie Cellulaire et Thérapeutique U944, INSERM, CNRS, Paris, France
| | - Frank Ling
- Université de Paris, Génomes, Biologie Cellulaire et Thérapeutique U944, INSERM, CNRS, Paris, France
| | - Gaetano Sodaro
- Université de Paris, Génomes, Biologie Cellulaire et Thérapeutique U944, INSERM, CNRS, Paris, France
| | - Paul-Arthur Meslin
- Université de Paris, Génomes, Biologie Cellulaire et Thérapeutique U944, INSERM, CNRS, Paris, France
| | | | - Nina Fenouille
- Université de Paris, Génomes, Biologie Cellulaire et Thérapeutique U944, INSERM, CNRS, Paris, France
| | - Jacob Hoj
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC, USA
| | - Rachel Washart
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC, USA
| | - Hazel X Ang
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC, USA
| | | | | | | | | | - Antoine Forget
- Université de Paris, Génomes, Biologie Cellulaire et Thérapeutique U944, INSERM, CNRS, Paris, France
| | - Raphael Itzykson
- Université de Paris, Génomes, Biologie Cellulaire et Thérapeutique U944, INSERM, CNRS, Paris, France
| | - Min Lu
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC, USA
| | - Jiaxing Lin
- Department of Biostatistics and Bioinformatics, Duke University, Durham, NC, USA
| | - Mariaelena Pierobon
- Center for Applied Proteomics and Molecular Medicine, School of Systems Biology, George Mason University, Manassas, VA, USA
| | - Zhecheng Sheng
- Department of Biostatistics and Bioinformatics, Duke University, Durham, NC, USA
| | - Xinghai Li
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Ashutosh Chilkoti
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Kouros Owzar
- Department of Biostatistics and Bioinformatics, Duke University, Durham, NC, USA
| | - David A Rizzieri
- Department of Medicine, Duke University Medical Center, Durham, NC, USA
| | - Timothy S Pardee
- Department of Internal Medicine, Section on Hematology and Oncology, Wake Forest Baptist Health, Winston-Salem, NC, USA
| | - Lina Benajiba
- Université de Paris, Génomes, Biologie Cellulaire et Thérapeutique U944, INSERM, CNRS, Paris, France
| | - Emanuel Petricoin
- Center for Applied Proteomics and Molecular Medicine, School of Systems Biology, George Mason University, Manassas, VA, USA
| | - Alexandre Puissant
- Université de Paris, Génomes, Biologie Cellulaire et Thérapeutique U944, INSERM, CNRS, Paris, France.
| | - Kris C Wood
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC, USA.
| |
Collapse
|
10
|
Yunes SA, Willoughby JLS, Kwan JH, Biagi JM, Pokharel N, Chin HG, York EA, Su KC, George K, Shah JV, Emili A, Schaus SE, Hansen U. Factor quinolinone inhibitors disrupt spindles and multiple LSF (TFCP2)-protein interactions in mitosis, including with microtubule-associated proteins. PLoS One 2022; 17:e0268857. [PMID: 35704642 PMCID: PMC9200292 DOI: 10.1371/journal.pone.0268857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 05/09/2022] [Indexed: 11/19/2022] Open
Abstract
Factor quinolinone inhibitors (FQIs), a first-in-class set of small molecule inhibitors targeted to the transcription factor LSF (TFCP2), exhibit promising cancer chemotherapeutic properties. FQI1, the initial lead compound identified, unexpectedly induced a concentration-dependent delay in mitotic progression. Here, we show that FQI1 can rapidly and reversibly lead to mitotic arrest, even when added directly to mitotic cells, implying that FQI1-mediated mitotic defects are not transcriptionally based. Furthermore, treatment with FQIs resulted in a striking, concentration-dependent diminishment of spindle microtubules, accompanied by a concentration-dependent increase in multi-aster formation. Aberrant γ-tubulin localization was also observed. These phenotypes suggest that perturbation of spindle microtubules is the primary event leading to the mitotic delays upon FQI1 treatment. Previously, FQIs were shown to specifically inhibit not only LSF DNA-binding activity, which requires LSF oligomerization to tetramers, but also other specific LSF-protein interactions. Other transcription factors participate in mitosis through non-transcriptional means, and we recently reported that LSF directly binds α-tubulin and is present in purified cellular tubulin preparations. Consistent with a microtubule role for LSF, here we show that LSF enhanced the rate of tubulin polymerization in vitro, and FQI1 inhibited such polymerization. To probe whether the FQI1-mediated spindle abnormalities could result from inhibition of mitotic LSF-protein interactions, mass spectrometry was performed using as bait an inducible, tagged form of LSF that is biotinylated by endogenous enzymes. The global proteomics analysis yielded expected associations for a transcription factor, notably with RNA processing machinery, but also to nontranscriptional components. In particular, and consistent with spindle disruption due to FQI treatment, mitotic, FQI1-sensitive interactions were identified between the biotinylated LSF and microtubule-associated proteins that regulate spindle assembly, positioning, and dynamics, as well as centrosome-associated proteins. Probing the mitotic LSF interactome using small molecule inhibitors therefore supported a non-transcriptional role for LSF in mediating progression through mitosis.
Collapse
Affiliation(s)
- Sarah A. Yunes
- Department of Biology, Boston University, Boston, Massachusetts, United States of America
- Program in Molecular Biology, Cell Biology, and Biochemistry, Boston University, Boston, Massachusetts, United States of America
| | - Jennifer L. S. Willoughby
- Program in Molecular Biology, Cell Biology, and Biochemistry, Boston University, Boston, Massachusetts, United States of America
- Alnylam Pharmaceuticals, Cambridge, Massachusetts, United States of America
| | - Julian H. Kwan
- Department of Biochemistry and Center for Network Systems Biology, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Jessica M. Biagi
- Department of Chemistry and Center for Molecular Discovery, Boston University, Boston, Massachusetts, United States of America
| | - Niranjana Pokharel
- Department of Chemistry and Center for Molecular Discovery, Boston University, Boston, Massachusetts, United States of America
| | - Hang Gyeong Chin
- Program in Molecular Biology, Cell Biology, and Biochemistry, Boston University, Boston, Massachusetts, United States of America
- New England Biolabs, Ipswich, Massachusetts, United States of America
| | - Emily A. York
- Department of Chemistry and Center for Molecular Discovery, Boston University, Boston, Massachusetts, United States of America
| | - Kuan-Chung Su
- Whitehead Institute for Biomedical Research, Cambridge, Massachusetts, United States of America
| | - Kelly George
- Department of Systems Biology, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Jagesh V. Shah
- Department of Systems Biology, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Andrew Emili
- Department of Biology, Boston University, Boston, Massachusetts, United States of America
- Department of Biochemistry and Center for Network Systems Biology, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Scott E. Schaus
- Department of Chemistry and Center for Molecular Discovery, Boston University, Boston, Massachusetts, United States of America
| | - Ulla Hansen
- Department of Biology, Boston University, Boston, Massachusetts, United States of America
- Program in Molecular Biology, Cell Biology, and Biochemistry, Boston University, Boston, Massachusetts, United States of America
| |
Collapse
|
11
|
Lagadec F, Carlon-Andres I, Ragues J, Port S, Wodrich H, Kehlenbach RH. CRM1 Promotes Capsid Disassembly and Nuclear Envelope Translocation of Adenovirus Independently of Its Export Function. J Virol 2022; 96:e0127321. [PMID: 34757845 PMCID: PMC8826800 DOI: 10.1128/jvi.01273-21] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 11/01/2021] [Indexed: 11/20/2022] Open
Abstract
After receptor-mediated endocytosis and endosomal escape, adenoviral capsids can travel via microtubule organizing centers to the nuclear envelope. Upon capsid disassembly, viral genome import into nuclei of interphase cells then occurs through nuclear pore complexes, involving the nucleoporins Nup214 and Nup358. Import also requires the activity of the classic nuclear export receptor CRM1, as it is blocked by the selective inhibitor leptomycin B. We have now used artificially enucleated as well as mitotic cells to analyze the role of an intact nucleus in different steps of the viral life cycle. In enucleated U2OS cells, viral capsids traveled to the microtubule organizing center, whereas their removal from this complex was blocked, suggesting that this step required nuclear factors. In mitotic cells, on the other hand, CRM1 promoted capsid disassembly and genome release, suggesting a role of this protein that does not require intact nuclear envelopes or nuclear pore complexes and is distinct from its function as a nuclear export receptor. Similar to enucleation, inhibition of CRM1 by leptomycin B also leads to an arrest of adenoviral capsids at the microtubule organizing center. In a small-scale screen using leptomycin B-resistant versions of CRM1, we identified a mutant, CRM1 W142A P143A, that is compromised with respect to adenoviral capsid disassembly in both interphase and mitotic cells. Strikingly, this mutant is capable of exporting cargo proteins out of the nucleus of living cells or digitonin-permeabilized cells, pointing to a role of the mutated region that is not directly linked to nuclear export. IMPORTANCE A role of nucleoporins and of soluble transport factors in adenoviral genome import into the nucleus of infected cells in interphase has previously been established. The nuclear export receptor CRM1 promotes genome import, but its precise function is not known. Using enucleated and mitotic cells, we showed that CRM1 does not simply function by exporting a crucial factor out of the nucleus that would then trigger capsid disassembly and genome import. Instead, CRM1 has an export-independent role, a notion that is also supported by a mutant, CRM1 W142A P143A, which is export competent but deficient in viral capsid disassembly, in both interphase and mitotic cells.
Collapse
Affiliation(s)
- Floriane Lagadec
- Department of Molecular Biology, Faculty of Medicine, Göttingen Center of Biosciences (GZMB), Georg-August-University Göttingen, Göttingen, Germany
- CNRS UMR 5234, Fundamental Microbiology and Pathogenicity, Université de Bordeaux, Bordeaux, France
| | - Irene Carlon-Andres
- CNRS UMR 5234, Fundamental Microbiology and Pathogenicity, Université de Bordeaux, Bordeaux, France
| | - Jessica Ragues
- CNRS UMR 5234, Fundamental Microbiology and Pathogenicity, Université de Bordeaux, Bordeaux, France
| | - Sarah Port
- Department of Molecular Biology, Faculty of Medicine, Göttingen Center of Biosciences (GZMB), Georg-August-University Göttingen, Göttingen, Germany
| | - Harald Wodrich
- CNRS UMR 5234, Fundamental Microbiology and Pathogenicity, Université de Bordeaux, Bordeaux, France
| | - Ralph H. Kehlenbach
- Department of Molecular Biology, Faculty of Medicine, Göttingen Center of Biosciences (GZMB), Georg-August-University Göttingen, Göttingen, Germany
| |
Collapse
|
12
|
Galinski B, Alexander TB, Mitchell DA, Chatwin HV, Awah C, Green AL, Weiser DA. Therapeutic Targeting of Exportin-1 in Childhood Cancer. Cancers (Basel) 2021; 13:6161. [PMID: 34944778 PMCID: PMC8699059 DOI: 10.3390/cancers13246161] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 11/16/2021] [Accepted: 12/01/2021] [Indexed: 01/24/2023] Open
Abstract
Overexpression of Exportin-1 (XPO1), a key regulator of nuclear-to-cytoplasmic transport, is associated with inferior patient outcomes across a range of adult malignancies. Targeting XPO1 with selinexor has demonstrated promising results in clinical trials, leading to FDA approval of its use for multiple relapsed/refractory cancers. However, XPO1 biology and selinexor sensitivity in childhood cancer is only recently being explored. In this review, we will focus on the differential biology of childhood and adult cancers as it relates to XPO1 and key cargo proteins. We will further explore the current state of pre-clinical and clinical development of XPO1 inhibitors in childhood cancers. Finally, we will outline potentially promising future therapeutic strategies for, as well as potential challenges to, integrating XPO1 inhibition to improve outcomes for children with cancer.
Collapse
Affiliation(s)
- Basia Galinski
- Department of Pediatrics, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (B.G.); (D.A.M.); (C.A.)
| | - Thomas B. Alexander
- Department of Pediatrics, University of North Carolina, Chapel Hill, NC 27599, USA;
| | - Daniel A. Mitchell
- Department of Pediatrics, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (B.G.); (D.A.M.); (C.A.)
| | - Hannah V. Chatwin
- Morgan Adams Foundation Pediatric Brain Tumor Research Program, University of Colorado School of Medicine, Aurora, CO 80045, USA;
| | - Chidiebere Awah
- Department of Pediatrics, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (B.G.); (D.A.M.); (C.A.)
| | - Adam L. Green
- Morgan Adams Foundation Pediatric Brain Tumor Research Program, University of Colorado School of Medicine, Aurora, CO 80045, USA;
| | - Daniel A. Weiser
- Department of Pediatrics, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (B.G.); (D.A.M.); (C.A.)
| |
Collapse
|
13
|
Nord MS, Bernis C, Carmona S, Garland DC, Travesa A, Forbes DJ. Exportins can inhibit major mitotic assembly events in vitro: membrane fusion, nuclear pore formation, and spindle assembly. Nucleus 2021; 11:178-193. [PMID: 32762441 PMCID: PMC7540616 DOI: 10.1080/19491034.2020.1798093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Xenopus egg extracts are a powerful in vitro tool for studying complex biological processes, including nuclear reconstitution, nuclear membrane and pore assembly, and spindle assembly. Extracts have been further used to demonstrate a moonlighting regulatory role for nuclear import receptors or importins on these cell cycle assembly events. Here we show that exportins can also play a role in these events. Addition of Crm1, Exportin-t, or Exportin-5 decreased nuclear pore assembly in vitro. RanQ69L-GTP, a constitutively active form of RanGTP, ameliorated inhibition. Both Crm1 and Exportin-t inhibited fusion of nuclear membranes, again counteracted by RanQ69L-GTP. In mitotic extracts, Crm1 and Exportin-t negatively impacted spindle assembly. Pulldowns from the extracts using Crm1- or Exportin-t-beads revealed nucleoporins known to be essential for both nuclear pore and spindle assembly, with RanQ69L-GTP decreasing a subset of these target interactions. This study suggests a model where exportins, like importins, can regulate major mitotic assembly events.
Collapse
Affiliation(s)
- Matthew S Nord
- Section of Cell and Developmental Biology, Division of Biological Sciences 0347, University of California-San Diego , La Jolla, CA, USA
| | - Cyril Bernis
- Section of Cell and Developmental Biology, Division of Biological Sciences 0347, University of California-San Diego , La Jolla, CA, USA
| | - Sarah Carmona
- Section of Cell and Developmental Biology, Division of Biological Sciences 0347, University of California-San Diego , La Jolla, CA, USA
| | - Dennis C Garland
- Section of Cell and Developmental Biology, Division of Biological Sciences 0347, University of California-San Diego , La Jolla, CA, USA
| | - Anna Travesa
- Section of Cell and Developmental Biology, Division of Biological Sciences 0347, University of California-San Diego , La Jolla, CA, USA
| | - Douglass J Forbes
- Section of Cell and Developmental Biology, Division of Biological Sciences 0347, University of California-San Diego , La Jolla, CA, USA
| |
Collapse
|
14
|
Bloomfield M, Chen J, Cimini D. Spindle Architectural Features Must Be Considered Along With Cell Size to Explain the Timing of Mitotic Checkpoint Silencing. Front Physiol 2021; 11:596263. [PMID: 33584330 PMCID: PMC7877541 DOI: 10.3389/fphys.2020.596263] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 12/23/2020] [Indexed: 11/25/2022] Open
Abstract
Mitosis proceeds through a defined series of events that is largely conserved, but the amount of time needed for their completion can vary in different cells and organisms. In many systems, mitotic duration depends on the time required to satisfy and silence the spindle assembly checkpoint (SAC), also known as the mitotic checkpoint. Because SAC silencing involves trafficking SAC molecules among kinetochores, spindle, and cytoplasm, the size and geometry of the spindle relative to cell volume are expected to affect mitotic duration by influencing the timing of SAC silencing. However, the relationship between SAC silencing, cell size, and spindle dimensions is unclear. To investigate this issue, we used four DLD-1 tetraploid (4N) clones characterized by small or large nuclear and cell size. We found that the small 4N clones had longer mitotic durations than the parental DLD-1 cells and that this delay was due to differences in their metaphase duration. Leveraging a previous mathematical model for spatiotemporal regulation of SAC silencing, we show that the difference in metaphase duration, i.e., SAC silencing time, can be explained by the distinct spindle microtubule densities and sizes of the cell, spindle, and spindle poles in the 4N clones. Lastly, we demonstrate that manipulating spindle geometry can alter mitotic and metaphase duration, consistent with a model prediction. Our results suggest that spindle size does not always scale with cell size in mammalian cells and cell size is not sufficient to explain the differences in metaphase duration. Only when a number of spindle architectural features are considered along with cell size can the kinetics of SAC silencing, and hence mitotic duration, in the different clones be explained.
Collapse
Affiliation(s)
- Mathew Bloomfield
- Department of Biological Sciences and Fralin Life Sciences Institute, Virginia Tech, Blacksburg, VA, United States
| | - Jing Chen
- Department of Biological Sciences and Fralin Life Sciences Institute, Virginia Tech, Blacksburg, VA, United States
| | - Daniela Cimini
- Department of Biological Sciences and Fralin Life Sciences Institute, Virginia Tech, Blacksburg, VA, United States
| |
Collapse
|
15
|
Verbeke D, Demeyer S, Prieto C, de Bock CE, De Bie J, Gielen O, Jacobs K, Mentens N, Verhoeven BM, Uyttebroeck A, Boeckx N, De Keersmaecker K, Maertens J, Segers H, Cools J. The XPO1 Inhibitor KPT-8602 Synergizes with Dexamethasone in Acute Lymphoblastic Leukemia. Clin Cancer Res 2020; 26:5747-5758. [PMID: 32826328 DOI: 10.1158/1078-0432.ccr-20-1315] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 07/20/2020] [Accepted: 08/18/2020] [Indexed: 11/16/2022]
Abstract
PURPOSE KPT-8602 (Eltanexor) is a second-generation exportin-1 (XPO1) inhibitor with potent activity against acute lymphoblastic leukemia (ALL) in preclinical models and with minimal effects on normal cells. In this study, we evaluated whether KPT-8602 would synergize with dexamethasone, vincristine, or doxorubicin, three drugs currently used for the treatment of ALL. EXPERIMENTAL DESIGN First, we searched for the most synergistic combination of KPT-8602 with dexamethasone, vincristine, or doxorubicin in vitro in both B-ALL and T-ALL cell lines using proliferation and apoptosis as a readout. Next, we validated this synergistic effect by treatment of clinically relevant B- and T-ALL patient-derived xenograft models in vivo. Finally, we performed RNA-sequencing (RNA-seq) and chromatin immunoprecipitation sequencing (ChIP-seq) to determine the mechanism of synergy. RESULTS KPT-8602 showed strong synergism with dexamethasone on human B-ALL and T-ALL cell lines as well as in vivo in three patient-derived ALL xenografts. Compared with single-drug treatment, the drug combination caused increased apoptosis and led to histone depletion. Mechanistically, integration of ChIP-seq and RNA-seq data revealed that addition of KPT-8602 to dexamethasone enhanced the activity of the glucocorticoid receptor (NR3C1) and led to increased inhibition of E2F-mediated transcription. We observed strong inhibition of E2F target genes related to cell cycle, DNA replication, and transcriptional regulation. CONCLUSIONS Our preclinical study demonstrates that KPT-8602 enhances the effects of dexamethasone to inhibit B-ALL and T-ALL cells via NR3C1- and E2F-mediated transcriptional complexes, allowing to achieve increased dexamethasone effects for patients.
Collapse
Affiliation(s)
- Delphine Verbeke
- Center for Human Genetics, KU Leuven, Leuven, Belgium
- Center for Cancer Biology, VIB, Leuven, Belgium
- Leuvens Kanker Instituut (LKI), KU Leuven - UZ Leuven, Leuven, Belgium
| | - Sofie Demeyer
- Center for Human Genetics, KU Leuven, Leuven, Belgium
- Center for Cancer Biology, VIB, Leuven, Belgium
- Leuvens Kanker Instituut (LKI), KU Leuven - UZ Leuven, Leuven, Belgium
| | - Cristina Prieto
- Center for Human Genetics, KU Leuven, Leuven, Belgium
- Center for Cancer Biology, VIB, Leuven, Belgium
- Leuvens Kanker Instituut (LKI), KU Leuven - UZ Leuven, Leuven, Belgium
| | - Charles E de Bock
- Children's Cancer Institute, Lowy Cancer Research Centre, University of New South Wales, Randwick, New South Wales, Australia
| | - Jolien De Bie
- Center for Human Genetics, KU Leuven, Leuven, Belgium
- Center for Cancer Biology, VIB, Leuven, Belgium
- Leuvens Kanker Instituut (LKI), KU Leuven - UZ Leuven, Leuven, Belgium
- Department of Laboratory Medicine, UZ Leuven, Leuven, Belgium
| | - Olga Gielen
- Center for Human Genetics, KU Leuven, Leuven, Belgium
- Center for Cancer Biology, VIB, Leuven, Belgium
- Leuvens Kanker Instituut (LKI), KU Leuven - UZ Leuven, Leuven, Belgium
| | - Kris Jacobs
- Center for Human Genetics, KU Leuven, Leuven, Belgium
- Center for Cancer Biology, VIB, Leuven, Belgium
- Leuvens Kanker Instituut (LKI), KU Leuven - UZ Leuven, Leuven, Belgium
| | - Nicole Mentens
- Center for Human Genetics, KU Leuven, Leuven, Belgium
- Center for Cancer Biology, VIB, Leuven, Belgium
- Leuvens Kanker Instituut (LKI), KU Leuven - UZ Leuven, Leuven, Belgium
| | - Bronte Manouk Verhoeven
- Center for Human Genetics, KU Leuven, Leuven, Belgium
- Center for Cancer Biology, VIB, Leuven, Belgium
- Leuvens Kanker Instituut (LKI), KU Leuven - UZ Leuven, Leuven, Belgium
| | - Anne Uyttebroeck
- Leuvens Kanker Instituut (LKI), KU Leuven - UZ Leuven, Leuven, Belgium
- Department of Pediatric Oncology, UZ Leuven, Leuven, Belgium
| | - Nancy Boeckx
- Department of Laboratory Medicine, UZ Leuven, Leuven, Belgium
- Department of Oncology, KU Leuven, Leuven, Belgium
| | - Kim De Keersmaecker
- Leuvens Kanker Instituut (LKI), KU Leuven - UZ Leuven, Leuven, Belgium
- Department of Oncology, KU Leuven, Leuven, Belgium
| | - Johan Maertens
- Leuvens Kanker Instituut (LKI), KU Leuven - UZ Leuven, Leuven, Belgium
- Department of Hematology, UZ Leuven, Leuven, Belgium
| | - Heidi Segers
- Leuvens Kanker Instituut (LKI), KU Leuven - UZ Leuven, Leuven, Belgium
- Department of Pediatric Oncology, UZ Leuven, Leuven, Belgium
| | - Jan Cools
- Center for Human Genetics, KU Leuven, Leuven, Belgium.
- Center for Cancer Biology, VIB, Leuven, Belgium
- Leuvens Kanker Instituut (LKI), KU Leuven - UZ Leuven, Leuven, Belgium
| |
Collapse
|
16
|
Gales JP, Kubina J, Geldreich A, Dimitrova M. Strength in Diversity: Nuclear Export of Viral RNAs. Viruses 2020; 12:E1014. [PMID: 32932882 PMCID: PMC7551171 DOI: 10.3390/v12091014] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 09/03/2020] [Accepted: 09/09/2020] [Indexed: 12/11/2022] Open
Abstract
The nuclear export of cellular mRNAs is a complex process that requires the orchestrated participation of many proteins that are recruited during the early steps of mRNA synthesis and processing. This strategy allows the cell to guarantee the conformity of the messengers accessing the cytoplasm and the translation machinery. Most transcripts are exported by the exportin dimer Nuclear RNA export factor 1 (NXF1)-NTF2-related export protein 1 (NXT1) and the transcription-export complex 1 (TREX1). Some mRNAs that do not possess all the common messenger characteristics use either variants of the NXF1-NXT1 pathway or CRM1, a different exportin. Viruses whose mRNAs are synthesized in the nucleus (retroviruses, the vast majority of DNA viruses, and influenza viruses) exploit both these cellular export pathways. Viral mRNAs hijack the cellular export machinery via complex secondary structures recognized by cellular export factors and/or viral adapter proteins. This way, the viral transcripts succeed in escaping the host surveillance system and are efficiently exported for translation, allowing the infectious cycle to proceed. This review gives an overview of the cellular mRNA nuclear export mechanisms and presents detailed insights into the most important strategies that viruses use to export the different forms of their RNAs from the nucleus to the cytoplasm.
Collapse
Affiliation(s)
- Jón Pol Gales
- Institut de Biologie Moléculaire des Plantes, The French National Center for Scientific Research (CNRS) UPR2357, Université de Strasbourg, F-67084 Strasbourg, France; (J.P.G.); (J.K.); (A.G.)
| | - Julie Kubina
- Institut de Biologie Moléculaire des Plantes, The French National Center for Scientific Research (CNRS) UPR2357, Université de Strasbourg, F-67084 Strasbourg, France; (J.P.G.); (J.K.); (A.G.)
- SVQV UMR-A 1131, INRAE, Université de Strasbourg, F-68000 Colmar, France
| | - Angèle Geldreich
- Institut de Biologie Moléculaire des Plantes, The French National Center for Scientific Research (CNRS) UPR2357, Université de Strasbourg, F-67084 Strasbourg, France; (J.P.G.); (J.K.); (A.G.)
| | - Maria Dimitrova
- Institut de Biologie Moléculaire des Plantes, The French National Center for Scientific Research (CNRS) UPR2357, Université de Strasbourg, F-67084 Strasbourg, France; (J.P.G.); (J.K.); (A.G.)
| |
Collapse
|
17
|
Cunha-Silva S, Conde C. From the Nuclear Pore to the Fibrous Corona: A MAD Journey to Preserve Genome Stability. Bioessays 2020; 42:e2000132. [PMID: 32885448 DOI: 10.1002/bies.202000132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Revised: 07/22/2020] [Indexed: 11/09/2022]
Abstract
The relationship between kinetochores and nuclear pore complexes (NPCs) is intimate but poorly understood. Several NPC components and associated proteins are relocated to mitotic kinetochores to assist in different activities that ensure faithful chromosome segregation. Such is the case of the Mad1-c-Mad2 complex, the catalytic core of the spindle assembly checkpoint (SAC), a surveillance pathway that delays anaphase until all kinetochores are attached to spindle microtubules. Mad1-c-Mad2 is recruited to discrete domains of unattached kinetochores from where it promotes the rate-limiting step in the assembly of anaphase-inhibitory complexes. SAC proficiency further requires Mad1-c-Mad2 to be anchored at NPCs during interphase. However, the mechanistic relevance of this arrangement for SAC function remains ill-defined. Recent studies uncover the molecular underpinnings that coordinate the release of Mad1-c-Mad2 from NPCs with its prompt recruitment to kinetochores. Here, current knowledge on Mad1-c-Mad2 function and spatiotemporal regulation is reviewed and the critical questions that remain unanswered are highlighted.
Collapse
Affiliation(s)
- Sofia Cunha-Silva
- i3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, 4200-135, Portugal.,IBMC, Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, 4200-135, Portugal.,Programa Doutoral em Biologia Molecular e Celular (MCbiology), Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, Porto, 4050-313, Portugal
| | - Carlos Conde
- i3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, 4200-135, Portugal.,IBMC, Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, 4200-135, Portugal
| |
Collapse
|
18
|
Baumhardt JM, Walker JS, Lee Y, Shakya B, Brautigam CA, Lapalombella R, Grishin N, Chook YM. Recognition of nuclear export signals by CRM1 carrying the oncogenic E571K mutation. Mol Biol Cell 2020; 31:1879-1891. [PMID: 32520643 PMCID: PMC7525811 DOI: 10.1091/mbc.e20-04-0233] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 05/26/2020] [Accepted: 06/04/2020] [Indexed: 12/28/2022] Open
Abstract
The E571K mutation of CRM1 is highly prevalent in some cancers, but its mechanism of tumorigenesis is unclear. Glu571 of CRM1 is located in its nuclear export signal (NES)-binding groove, suggesting that binding of select NESs may be altered. We generated HEK 293 cells with either monoallelic CRM1WT/E571K or biallelic CRM1E571K/E571K using CRISPR/Cas9. We also combined analysis of binding affinities and structures of 27 diverse NESs for wild-type and E571K CRM1 with structure-based bioinformatics. While most NESs bind the two CRM1 similarly, NESs from Mek1, eIF4E-transporter, and RPS2 showed >10-fold affinity differences. These NESs have multiple charged side chains binding close to CRM1 position 571, but this feature alone was not sufficient to predict different binding to CRM1(E571K). Consistent with eIF4E-transporter NES binding weaker to CRM1(E571K), eIF4E-transporter was mislocalized in tumor cells carrying CRM1(E571K). This serves as proof of concept that understanding how CRM1(E571K) affects NES binding provides a platform for identifying cargoes that are mislocalized in cancer upon CRM1 mutation. Finally, we showed that large affinity changes seen with some NES peptides (of Mek1 and RPS2) do not always translate to the full-length cargoes, suggesting limitations with current NES prediction methods. Therefore, comprehensive studies like ours are imperative to identify CRM1 cargoes with real pathogenic potential.
Collapse
Affiliation(s)
- Jordan M. Baumhardt
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Janek S. Walker
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, OH 43210
| | - Yoonji Lee
- Departments of Biophysics and Microbiology and Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Binita Shakya
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Chad A. Brautigam
- Departments of Biophysics and Microbiology and Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Rosa Lapalombella
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, OH 43210
| | - Nick Grishin
- Departments of Biophysics and Microbiology and Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Yuh Min Chook
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX 75390
| |
Collapse
|
19
|
Li X, Feng Y, Yan M, Tu X, Xie B, Ni F, Qu C, Chen JG. Inhibition of Autism-Related Crm1 Disrupts Mitosis and Induces Apoptosis of the Cortical Neural Progenitors. Cereb Cortex 2020; 30:3960-3976. [PMID: 32008040 DOI: 10.1093/cercor/bhaa011] [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: 10/08/2019] [Revised: 12/06/2019] [Accepted: 01/11/2020] [Indexed: 11/14/2022] Open
Abstract
De novo microdeletion of chromosome 2p15-16.1 presents clinically recognizable phenotypes that include mental retardation, autism, and microcephaly. Chromosomal maintenance 1 (CRM1) is a gene commonly missing in patients with 2p15-16.1 microdeletion and one of two genes found in the smallest deletion case. In this study, we investigate the role and mechanism of Crm1 in the developing mouse brain by inhibiting the protein or knocking down the gene in vivo. Inhibition of Crm1 reduces the proliferation and increases p53-dependent apoptosis of the cortical neural progenitors, thereby impeding the growth of embryonic cerebral cortex. Live imaging of mitosis in ex vivo embryonic brain slices reveals that inhibition of CRM1 arrests the cortical progenitors at metaphase. The arrested cells eventually slip into a pseudo-G1 phase without chromosome segregation. The mitotic slippage cells are marked by persistent expression of the spindle assembly checkpoint (SAC), repressing of which rescues the cells from apoptosis. Our study reveals that activating the SAC and inducing the mitotic slippage may lead to apoptosis of the cortical neural progenitors. The resulting cell death may well contribute to microcephaly associated with microdeletion of chromosome 2p15-16.1 involving CRM1.
Collapse
Affiliation(s)
- Xue Li
- School of Ophthalmology and Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325027, P.R. China
- State Key Laboratory of Optometry, Ophthalmology and Vision Science and Zhejiang Provincial Key Laboratory of Optometry and Ophthalmology, Wenzhou, Zhejiang 325027, P.R. China
| | - Yue Feng
- School of Ophthalmology and Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325027, P.R. China
- State Key Laboratory of Optometry, Ophthalmology and Vision Science and Zhejiang Provincial Key Laboratory of Optometry and Ophthalmology, Wenzhou, Zhejiang 325027, P.R. China
| | - Meifang Yan
- School of Ophthalmology and Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325027, P.R. China
- State Key Laboratory of Optometry, Ophthalmology and Vision Science and Zhejiang Provincial Key Laboratory of Optometry and Ophthalmology, Wenzhou, Zhejiang 325027, P.R. China
| | - Xiaomeng Tu
- School of Ophthalmology and Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325027, P.R. China
- State Key Laboratory of Optometry, Ophthalmology and Vision Science and Zhejiang Provincial Key Laboratory of Optometry and Ophthalmology, Wenzhou, Zhejiang 325027, P.R. China
| | - Bin Xie
- School of Ophthalmology and Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325027, P.R. China
- State Key Laboratory of Optometry, Ophthalmology and Vision Science and Zhejiang Provincial Key Laboratory of Optometry and Ophthalmology, Wenzhou, Zhejiang 325027, P.R. China
| | - Fangfang Ni
- School of Ophthalmology and Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325027, P.R. China
- State Key Laboratory of Optometry, Ophthalmology and Vision Science and Zhejiang Provincial Key Laboratory of Optometry and Ophthalmology, Wenzhou, Zhejiang 325027, P.R. China
| | - Chunsheng Qu
- Clinical Laboratory of Lishui People's Hospital, Sixth Affiliated Hospital, Wenzhou Medical University, LiShui, Zhejiang 323000, China
| | - Jie-Guang Chen
- School of Ophthalmology and Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325027, P.R. China
- State Key Laboratory of Optometry, Ophthalmology and Vision Science and Zhejiang Provincial Key Laboratory of Optometry and Ophthalmology, Wenzhou, Zhejiang 325027, P.R. China
| |
Collapse
|
20
|
Jackman M, Marcozzi C, Barbiero M, Pardo M, Yu L, Tyson AL, Choudhary JS, Pines J. Cyclin B1-Cdk1 facilitates MAD1 release from the nuclear pore to ensure a robust spindle checkpoint. J Cell Biol 2020; 219:e201907082. [PMID: 32236513 PMCID: PMC7265330 DOI: 10.1083/jcb.201907082] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 02/05/2020] [Accepted: 03/06/2020] [Indexed: 11/22/2022] Open
Abstract
How the cell rapidly and completely reorganizes its architecture when it divides is a problem that has fascinated researchers for almost 150 yr. We now know that the core regulatory machinery is highly conserved in eukaryotes, but how these multiple protein kinases, protein phosphatases, and ubiquitin ligases are coordinated in space and time to remodel the cell in a matter of minutes remains a major question. Cyclin B1-Cdk is the primary kinase that drives mitotic remodeling; here we show that it is targeted to the nuclear pore complex (NPC) by binding an acidic face of the kinetochore checkpoint protein, MAD1, where it coordinates NPC disassembly with kinetochore assembly. Localized cyclin B1-Cdk1 is needed for the proper release of MAD1 from the embrace of TPR at the nuclear pore so that it can be recruited to kinetochores before nuclear envelope breakdown to maintain genomic stability.
Collapse
|
21
|
Azizian NG, Li Y. XPO1-dependent nuclear export as a target for cancer therapy. J Hematol Oncol 2020; 13:61. [PMID: 32487143 PMCID: PMC7268335 DOI: 10.1186/s13045-020-00903-4] [Citation(s) in RCA: 110] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 05/22/2020] [Indexed: 01/08/2023] Open
Abstract
Cellular homeostasis requires the proper nuclear-cytoplasmic partitioning of large molecules, which is often deregulated in cancer. XPO1 is an export receptor responsible for the nuclear-cytoplasmic transport of hundreds of proteins and multiple RNA species. XPO1 is frequently overexpressed and/or mutated in human cancers and functions as an oncogenic driver. Suppression of XPO1-mediated nuclear export, therefore, presents a unique therapeutic strategy. In this review, we summarize the physiological functions of XPO1 as well as the development of various XPO1 inhibitors and provide an update on the recent clinical trials of the SINE compounds. We also discuss potential future research directions on the molecular function of XPO1 and the clinical application of XPO1 inhibitors.
Collapse
Affiliation(s)
- Nancy G Azizian
- Center for Immunotherapy Research, Houston Methodist Research Institute, 6670 Bertner Avenue, Houston, TX, 77030, USA
- Department of Medicine, Weill Cornell Medical College, New York, NY, 10065, USA
| | - Yulin Li
- Center for Immunotherapy Research, Houston Methodist Research Institute, 6670 Bertner Avenue, Houston, TX, 77030, USA.
- Department of Medicine, Weill Cornell Medical College, New York, NY, 10065, USA.
| |
Collapse
|
22
|
Bao XX, Spanos C, Kojidani T, Lynch EM, Rappsilber J, Hiraoka Y, Haraguchi T, Sawin KE. Exportin Crm1 is repurposed as a docking protein to generate microtubule organizing centers at the nuclear pore. eLife 2018; 7:e33465. [PMID: 29809148 PMCID: PMC6008054 DOI: 10.7554/elife.33465] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Accepted: 05/21/2018] [Indexed: 01/04/2023] Open
Abstract
Non-centrosomal microtubule organizing centers (MTOCs) are important for microtubule organization in many cell types. In fission yeast Schizosaccharomyces pombe, the protein Mto1, together with partner protein Mto2 (Mto1/2 complex), recruits the γ-tubulin complex to multiple non-centrosomal MTOCs, including the nuclear envelope (NE). Here, we develop a comparative-interactome mass spectrometry approach to determine how Mto1 localizes to the NE. Surprisingly, we find that Mto1, a constitutively cytoplasmic protein, docks at nuclear pore complexes (NPCs), via interaction with exportin Crm1 and cytoplasmic FG-nucleoporin Nup146. Although Mto1 is not a nuclear export cargo, it binds Crm1 via a nuclear export signal-like sequence, and docking requires both Ran in the GTP-bound state and Nup146 FG repeats. In addition to determining the mechanism of MTOC formation at the NE, our results reveal a novel role for Crm1 and the nuclear export machinery in the stable docking of a cytoplasmic protein complex at NPCs.
Collapse
Affiliation(s)
- Xun X Bao
- Wellcome Centre for Cell Biology, School of Biological SciencesUniversity of EdinburghEdinburghUnited Kingdom
| | - Christos Spanos
- Wellcome Centre for Cell Biology, School of Biological SciencesUniversity of EdinburghEdinburghUnited Kingdom
| | - Tomoko Kojidani
- Advanced ICT Research Institute KobeNational Institute of Information and Communications TechnologyKobeJapan
- Department of Chemical and Biological Sciences, Faculty of ScienceJapan Women’s UniversityTokyoJapan
| | - Eric M Lynch
- Wellcome Centre for Cell Biology, School of Biological SciencesUniversity of EdinburghEdinburghUnited Kingdom
| | - Juri Rappsilber
- Wellcome Centre for Cell Biology, School of Biological SciencesUniversity of EdinburghEdinburghUnited Kingdom
- Department of BioanalyticsInstitute of Biotechnology, Technische Universität BerlinBerlinGermany
| | - Yasushi Hiraoka
- Advanced ICT Research Institute KobeNational Institute of Information and Communications TechnologyKobeJapan
- Graduate School of Frontier BiosciencesOsaka UniversitySuitaJapan
| | - Tokuko Haraguchi
- Advanced ICT Research Institute KobeNational Institute of Information and Communications TechnologyKobeJapan
- Graduate School of Frontier BiosciencesOsaka UniversitySuitaJapan
| | - Kenneth E Sawin
- Wellcome Centre for Cell Biology, School of Biological SciencesUniversity of EdinburghEdinburghUnited Kingdom
| |
Collapse
|
23
|
Fu X, Liang C, Li F, Wang L, Wu X, Lu A, Xiao G, Zhang G. The Rules and Functions of Nucleocytoplasmic Shuttling Proteins. Int J Mol Sci 2018; 19:ijms19051445. [PMID: 29757215 PMCID: PMC5983729 DOI: 10.3390/ijms19051445] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Revised: 04/16/2018] [Accepted: 04/17/2018] [Indexed: 12/14/2022] Open
Abstract
Biological macromolecules are the basis of life activities. There is a separation of spatial dimension between DNA replication and RNA biogenesis, and protein synthesis, which is an interesting phenomenon. The former occurs in the cell nucleus, while the latter in the cytoplasm. The separation requires protein to transport across the nuclear envelope to realize a variety of biological functions. Nucleocytoplasmic transport of protein including import to the nucleus and export to the cytoplasm is a complicated process that requires involvement and interaction of many proteins. In recent years, many studies have found that proteins constantly shuttle between the cytoplasm and the nucleus. These shuttling proteins play a crucial role as transport carriers and signal transduction regulators within cells. In this review, we describe the mechanism of nucleocytoplasmic transport of shuttling proteins and summarize some important diseases related shuttling proteins.
Collapse
Affiliation(s)
- Xuekun Fu
- Department of Biology and Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Southern University of Science and Technology, Shenzhen 518055, China.
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China.
| | - Chao Liang
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China.
- Institute of Integrated Bioinfomedicine and Translational Science, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China.
- Institute of Precision Medicine and Innovative Drug Discovery, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China.
- Shenzhen Lab of Combinatorial Compounds and Targeted Drug Delivery, HKBU Institute of Research and Continuing Education, Shenzhen 518057, China.
| | - Fangfei Li
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China.
- Institute of Integrated Bioinfomedicine and Translational Science, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China.
- Institute of Precision Medicine and Innovative Drug Discovery, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China.
- Shenzhen Lab of Combinatorial Compounds and Targeted Drug Delivery, HKBU Institute of Research and Continuing Education, Shenzhen 518057, China.
| | - Luyao Wang
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China.
- Institute of Integrated Bioinfomedicine and Translational Science, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China.
- Institute of Precision Medicine and Innovative Drug Discovery, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China.
- Shenzhen Lab of Combinatorial Compounds and Targeted Drug Delivery, HKBU Institute of Research and Continuing Education, Shenzhen 518057, China.
| | - Xiaoqiu Wu
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China.
- Institute of Integrated Bioinfomedicine and Translational Science, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China.
- Institute of Precision Medicine and Innovative Drug Discovery, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China.
- Shenzhen Lab of Combinatorial Compounds and Targeted Drug Delivery, HKBU Institute of Research and Continuing Education, Shenzhen 518057, China.
| | - Aiping Lu
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China.
- Institute of Integrated Bioinfomedicine and Translational Science, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China.
- Institute of Precision Medicine and Innovative Drug Discovery, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China.
- Shenzhen Lab of Combinatorial Compounds and Targeted Drug Delivery, HKBU Institute of Research and Continuing Education, Shenzhen 518057, China.
| | - Guozhi Xiao
- Department of Biology and Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Southern University of Science and Technology, Shenzhen 518055, China.
- Department of Orthopedic Surgery, Rush University Medical Center, Chicago, IL 60612, USA.
| | - Ge Zhang
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China.
- Institute of Integrated Bioinfomedicine and Translational Science, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China.
- Institute of Precision Medicine and Innovative Drug Discovery, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China.
- Shenzhen Lab of Combinatorial Compounds and Targeted Drug Delivery, HKBU Institute of Research and Continuing Education, Shenzhen 518057, China.
| |
Collapse
|
24
|
Yue L, Sun ZN, Yao YS, Shen Z, Wang HB, Liu XP, Zhou F, Xiang JY, Yao RY, Niu HT. CRM1, a novel independent prognostic factor overexpressed in invasive breast carcinoma of poor prognosis. Oncol Lett 2018; 15:7515-7522. [PMID: 29725458 PMCID: PMC5920404 DOI: 10.3892/ol.2018.8316] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2015] [Accepted: 04/21/2017] [Indexed: 12/16/2022] Open
Abstract
Breast cancer (BC) is the most commonly diagnosed cancer in females globally and is more aggressive at later stages. Chromosome region maintenance 1 (CRM1) is involved in the nuclear export of proteins and RNAs and has been associated with a number of malignancies. However, the clinicopathological significance of its expression in BC remains to be elucidated therefore this was investigated in the present study. CRM1 expression in 280 breast cancer tissues and 60 normal tissues was retrospectively analyzed using immunohistochemistry (IHC) and western blotting. IHC investigation demonstrated that CRM1 expression was significantly increased in BC compared with the normal breast epithelium (P<0.0001). Overexpression of CRM1 was markedly associated with poor prognostic characteristics, including larger tumor size (P=0.024), positive lymph node metastasis (P=0.032), invasive histological type (P=0.004) and distant metastasis (P=0.026). Significant associations were also observed between increased CRM1 expression and the progesterone receptor (P=0.028) and Ki67 (P=0.019). Kaplan-Meier survival analysis demonstrated that patients with high CRM1 expression exhibited a reduced disease-free survival and overall survival compared with those with low CRM1 expression (P=0.013). In the multivariate analysis, CRM1 expression (P=0.011), tumor size (P=0.001) and lymph node metastasis (P<0.001) were independent prognostic markers of BC. In conclusion, CRM1 serves an important role in BC and may serve as a predictive and prognostic factor for a poor outcome in patients with BC.
Collapse
Affiliation(s)
- Lu Yue
- Department of Oncology, Qingdao Municipal Hospital, School of Medicine, Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Zhen-Ni Sun
- Department of Oncology, Qingdao Municipal Hospital, School of Medicine, Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Ya-Sai Yao
- Department of Oncology, Qingdao Municipal Hospital, School of Medicine, Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Zan Shen
- Department of Oncology, The Sixth People's Hospital, Medical College of Shanghai Jiao Tong University, Shanghai 200233, P.R. China
| | - Hai-Bo Wang
- Department of Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Xiang-Ping Liu
- Central Laboratory, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Fang Zhou
- Department of Radiotherapy, Yantai Yuhuangding Hospital Affiliated to Medical College of Qingdao University, Yantai, Shandong 264000, P.R. China
| | - Jin-Yu Xiang
- Department of Oncology, Yantai Yuhuangding Hospital Affiliated to Medical College of Qingdao University, Yantai, Shandong 264000, P.R. China
| | - Ru-Yong Yao
- Central Laboratory, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Hai-Tao Niu
- Department of Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| |
Collapse
|
25
|
Li J, Shima H, Nishizawa H, Ikeda M, Brydun A, Matsumoto M, Kato H, Saiki Y, Liu L, Watanabe-Matsui M, Iemura K, Tanaka K, Shiraki T, Igarashi K. Phosphorylation of BACH1 switches its function from transcription factor to mitotic chromosome regulator and promotes its interaction with HMMR. Biochem J 2018; 475:981-1002. [PMID: 29459360 DOI: 10.1042/bcj20170520] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Revised: 01/31/2018] [Accepted: 02/19/2018] [Indexed: 12/17/2023]
Abstract
The transcription repressor BACH1 performs mutually independent dual roles in transcription regulation and chromosome alignment during mitosis by supporting polar ejection force of mitotic spindle. We now found that the mitotic spindles became oblique relative to the adhesion surface following endogenous BACH1 depletion in HeLa cells. This spindle orientation rearrangement was rescued by re-expression of BACH1 depending on its interactions with HMMR and CRM1, both of which are required for the positioning of mitotic spindle, but independently of its DNA-binding activity. A mass spectrometry analysis of BACH1 complexes in interphase and M phase revealed that BACH1 lost during mitosis interactions with proteins involved in chromatin and gene expression but retained interactions with HMMR and its known partners including CHICA. By analyzing BACH1 modification using stable isotope labeling with amino acids in cell culture, mitosis-specific phosphorylations of BACH1 were observed, and mutations of these residues abolished the activity of BACH1 to restore mitotic spindle orientation in knockdown cells and to interact with HMMR. Detailed histological analysis of Bach1-deficient mice revealed lengthening of the epithelial fold structures of the intestine. These observations suggest that BACH1 performs stabilization of mitotic spindle orientation together with HMMR and CRM1 in mitosis, and that the cell cycle-specific phosphorylation switches the transcriptional and mitotic functions of BACH1.
Collapse
Affiliation(s)
- Jie Li
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan
| | - Hiroki Shima
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan
- Center for Regulatory Epigenome and Diseases, Tohoku University, Sendai 980-8575, Japan
| | - Hironari Nishizawa
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan
| | - Masatoshi Ikeda
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan
| | - Andrey Brydun
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan
| | - Mitsuyo Matsumoto
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan
- Center for Regulatory Epigenome and Diseases, Tohoku University, Sendai 980-8575, Japan
| | - Hiroki Kato
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan
| | - Yuriko Saiki
- Department of Pathology, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan
| | - Liang Liu
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan
| | - Miki Watanabe-Matsui
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan
- Restart Postdoctoral Fellow, Japan Society for the Promotion of Science, Tokyo 102-0083, Japan
| | - Kenji Iemura
- Department of Molecular Oncology, Institute of Development, Aging and Cancer, Tohoku University, Sendai 980-8575, Japan
| | - Kozo Tanaka
- Department of Molecular Oncology, Institute of Development, Aging and Cancer, Tohoku University, Sendai 980-8575, Japan
| | - Takuma Shiraki
- Department of Science and Technology on Food Safety, Faculty of Biology-Oriented Science and Technology, Kindai University, 930 Nishimitani, Kinokawashi 649-6493, Japan
| | - Kazuhiko Igarashi
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan
- Center for Regulatory Epigenome and Diseases, Tohoku University, Sendai 980-8575, Japan
| |
Collapse
|
26
|
RanGTPase regulates the interaction between the inner nuclear membrane proteins, Samp1 and Emerin. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2018; 1860:1326-1334. [PMID: 29510091 DOI: 10.1016/j.bbamem.2018.03.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Revised: 02/07/2018] [Accepted: 03/01/2018] [Indexed: 02/02/2023]
Abstract
Samp1, spindle associated membrane protein 1, is a type II integral membrane protein localized in the inner nuclear membrane. Recent studies have shown that the inner nuclear membrane protein, Emerin and the small monomeric GTPase, Ran are direct binding partners of Samp1. Here we addressed the question whether Ran could regulate the interaction between Samp1 and Emerin in the inner nuclear membrane. To investigate the interaction between Samp1 and Emerin in live cells, we performed FRAP experiments in cells overexpressing YFP-Emerin. We compared the mobility of YFP-Emerin in Samp1 knock out cells and cells overexpressing Samp1. The results showed that the mobility of YFP-Emerin was higher in Samp1 knock out cells and lower in cells overexpressing Samp1, suggesting that Samp1 significantly attenuates the mobility of Emerin in the nuclear envelope. FRAP experiments using tsBN2 cells showed that the mobility of Emerin depends on RanGTP. Consistently, in vitro binding experiments showed that the affinity between Samp1 and Emerin is decreased in the presence of Ran, suggesting that Ran attenuates the interaction between Samp1 and Emerin. This is the first demonstration that Ran can regulate the interaction between two proteins in the nuclear envelope.
Collapse
|
27
|
Gilistro E, de Turris V, Damizia M, Verrico A, Moroni S, De Santis R, Rosa A, Lavia P. Importin-β and CRM1 control a RANBP2 spatiotemporal switch essential for mitotic kinetochore function. J Cell Sci 2017; 130:2564-2578. [PMID: 28600321 DOI: 10.1242/jcs.197905] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Accepted: 06/05/2017] [Indexed: 12/30/2022] Open
Abstract
Protein conjugation with small ubiquitin-related modifier (SUMO) is a post-translational modification that modulates protein interactions and localisation. RANBP2 is a large nucleoporin endowed with SUMO E3 ligase and SUMO-stabilising activity, and is implicated in some cancer types. RANBP2 is part of a larger complex, consisting of SUMO-modified RANGAP1, the GTP-hydrolysis activating factor for the GTPase RAN. During mitosis, the RANBP2-SUMO-RANGAP1 complex localises to the mitotic spindle and to kinetochores after microtubule attachment. Here, we address the mechanisms that regulate this localisation and how they affect kinetochore functions. Using proximity ligation assays, we find that nuclear transport receptors importin-β and CRM1 play essential roles in localising the RANBP2-SUMO-RANGAP1 complex away from, or at kinetochores, respectively. Using newly generated inducible cell lines, we show that overexpression of nuclear transport receptors affects the timing of RANBP2 localisation in opposite ways. Concomitantly, kinetochore functions are also affected, including the accumulation of SUMO-conjugated topoisomerase-IIα and stability of kinetochore fibres. These results delineate a novel mechanism through which nuclear transport receptors govern the functional state of kinetochores by regulating the timely deposition of RANBP2.
Collapse
Affiliation(s)
- Eugenia Gilistro
- CNR National Research Council of Italy, Institute of Molecular Biology and Pathology (IBPM), ℅ Department of Biology and Biotechnology, Sapienza Università di Roma, Via degli Apuli 4, 00185 Rome, Italy
| | - Valeria de Turris
- Istituto Italiano di Tecnologia, Center for Life Nanoscience@Sapienza, Viale Regina Elena 291, 00161 Rome, Italy
| | - Michela Damizia
- CNR National Research Council of Italy, Institute of Molecular Biology and Pathology (IBPM), ℅ Department of Biology and Biotechnology, Sapienza Università di Roma, Via degli Apuli 4, 00185 Rome, Italy
| | - Annalisa Verrico
- CNR National Research Council of Italy, Institute of Molecular Biology and Pathology (IBPM), ℅ Department of Biology and Biotechnology, Sapienza Università di Roma, Via degli Apuli 4, 00185 Rome, Italy
| | - Sara Moroni
- CNR National Research Council of Italy, Institute of Molecular Biology and Pathology (IBPM), ℅ Department of Biology and Biotechnology, Sapienza Università di Roma, Via degli Apuli 4, 00185 Rome, Italy
| | - Riccardo De Santis
- Istituto Italiano di Tecnologia, Center for Life Nanoscience@Sapienza, Viale Regina Elena 291, 00161 Rome, Italy
- Department of Biology and Biotechnology 'Charles Darwin', Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy
| | - Alessandro Rosa
- Istituto Italiano di Tecnologia, Center for Life Nanoscience@Sapienza, Viale Regina Elena 291, 00161 Rome, Italy
- Department of Biology and Biotechnology 'Charles Darwin', Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy
| | - Patrizia Lavia
- CNR National Research Council of Italy, Institute of Molecular Biology and Pathology (IBPM), ℅ Department of Biology and Biotechnology, Sapienza Università di Roma, Via degli Apuli 4, 00185 Rome, Italy
| |
Collapse
|
28
|
Mathew C, Ghildyal R. CRM1 Inhibitors for Antiviral Therapy. Front Microbiol 2017; 8:1171. [PMID: 28702009 PMCID: PMC5487384 DOI: 10.3389/fmicb.2017.01171] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Accepted: 06/08/2017] [Indexed: 12/22/2022] Open
Abstract
Infectious diseases are a major global concern and despite major advancements in medical research, still cause significant morbidity and mortality. Progress in antiviral therapy is particularly hindered by appearance of mutants capable of overcoming the effects of drugs targeting viral components. Alternatively, development of drugs targeting host proteins essential for completion of viral lifecycle holds potential as a viable strategy for antiviral therapy. Nucleocytoplasmic trafficking pathways in particular are involved in several pathological conditions including cancer and viral infections, where hijacking or alteration of function of key transporter proteins, such as Chromosome Region Maintenance1 (CRM1) is observed. Overexpression of CRM1-mediated nuclear export is evident in several solid and hematological malignancies. Interestingly, CRM1-mediated nuclear export of viral components is crucial in various stages of the viral lifecycle and assembly. This review summarizes the role of CRM1 in cancer and selected viruses. Leptomycin B (LMB) is the prototypical inhibitor of CRM1 potent against various cancer cell lines overexpressing CRM1 and in limiting viral infections at nanomolar concentrations in vitro. However, the irreversible shutdown of nuclear export results in high cytotoxicity and limited efficacy in vivo. This has prompted search for synthetic and natural CRM1 inhibitors that can potentially be developed as broadly active antivirals, some of which are summarized in this review.
Collapse
Affiliation(s)
| | - Reena Ghildyal
- Respiratory Virology Group, Centre for Research in Therapeutic Solutions, Health Research Institute, University of CanberraCanberra, ACT, Australia
| |
Collapse
|
29
|
Vijayaraghavan B, Jafferali MH, Figueroa RA, Hallberg E. Samp1, a RanGTP binding transmembrane protein in the inner nuclear membrane. Nucleus 2017; 7:415-23. [PMID: 27541860 PMCID: PMC5039005 DOI: 10.1080/19491034.2016.1220465] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Samp1 is a transmembrane protein of the inner nuclear membrane (INM), which interacts with the nuclear lamina and the Linker of Nucleoskeleton and Cytoskeleton (LINC) complex in interphase and during mitosis, it localizes to the mitotic spindle. Samp1 was recently found to coprecipitate a protein complex containing Ran, a GTPase with fundamental regulatory functions both in interphase and in mitosis. To investigate the interaction between Samp1 and Ran in further detail, we have designed and expressed recombinant fusion proteins of the Chaetomium thermophilum homolog of Samp1 (Ct.Samp1) and human Ran. Pulldown experiments show that Samp1 binds directly to Ran and that Samp1 binds better to RanGTP compared to RanGDP. Samp1 also preferred RanGTP over RanGDP in living tsBN2 cells. We also show that the Ran binding domain is located between amino acids 75–135 in the nucleoplasmically exposed N-terminal tail of Samp1. This domain is unique for Samp1, without homology in any other proteins in fungi or metazoa. Samp1 is the first known transmembrane protein that binds to Ran and could provide a unique local binding site for RanGTP in the INM. Samp1 overexpression resulted in increased Ran concentrations in the nuclear periphery supporting this idea.
Collapse
Affiliation(s)
| | | | | | - Einar Hallberg
- a Department of Neurochemistry , Stockholm University , Stockholm , Sweden
| |
Collapse
|
30
|
Abstract
Mitosis is the stage of the cell cycle during which replicated chromosomes must be precisely divided to allow the formation of two daughter cells possessing equal genetic material. Much of the careful spatial and temporal organization of mitosis is maintained through post-translational modifications, such as phosphorylation and ubiquitination, of key cellular proteins. Here, we will review evidence that sumoylation, conjugation to the SUMO family of small ubiquitin-like modifiers, also serves essential regulatory roles during mitosis. We will discuss the basic biology of sumoylation, how the SUMO pathway has been implicated in particular mitotic functions, including chromosome condensation, centromere/kinetochore organization and cytokinesis, and what cellular proteins may be the targets underlying these phenomena.
Collapse
Affiliation(s)
- Debaditya Mukhopadhyay
- Section on Cell Cycle Regulation, Laboratory of Gene Regulation and Development, National Institute of Child Health and Development, National Institutes of Health, 18 Library Drive, Room 106, Building 18T, Bethesda, MD, 20892, USA
| | - Mary Dasso
- Section on Cell Cycle Regulation, Laboratory of Gene Regulation and Development, National Institute of Child Health and Development, National Institutes of Health, 18 Library Drive, Room 106, Building 18T, Bethesda, MD, 20892, USA.
| |
Collapse
|
31
|
Labarrade F, Botto JM, Domloge N. CRM1 and chromosomal passenger complex component survivin are essential to normal mitosis progress and to preserve keratinocytes from mitotic abnormalities. Int J Cosmet Sci 2016; 38:452-61. [PMID: 26859314 DOI: 10.1111/ics.12311] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Accepted: 02/04/2016] [Indexed: 12/14/2022]
Abstract
OBJECTIVE Human epidermis provides the body a barrier against environmental assaults. To assume this function, the epidermis needs the renewal of keratinocytes allowed by constant mitosis, which replace the exfoliating corneocytes. Keratinocyte stem cells (KSCs) located in the basal epidermis are mitotically active, self-renewing and govern the epithelial stratification by producing renewed source of keratinocytes. Protein complex such as the chromosomal passenger complex (CPC) allows the correct development of this process. The CPC is composed of four members: INCENP, survivin, borealin and aurora kinase B, and the disruption of the CPC during cell division induces mitotic spindle defects and improper repartition of chromosomes. The aim of our study was to investigate the implication of CRM1 and survivin in the progress of mitosis in skin keratinocytes. METHODS Cultured human keratinocytes and skin biopsies were used in this study. KSCs-enriched population of keratinocytes was isolated from total keratinocytes by differential attachment to a type IV collagen matrix. Survivin and CRM1 expression levels were assessed by immunofluorescence and immunoblotting. Specific siRNAs for each CPC member and for CRM1 were used to determine the relationship between these proteins. Survivin-specific siRNA was used to induce the apparition of mitotic abnormalities in cultured keratinocytes. RESULTS We demonstrated the ability of our compound 'IV08.009' to modulate the expression level of survivin and CRM1 in keratinocytes and in skin biopsies. We observed that members of the CPC are interdependent: siRNA-induced inhibition of one component caused a decrease in the expression of all other CPC members. Downregulation of survivin or CRM1 induced mitotic abnormalities in keratinocytes. However, decreased number of mitotic abnormalities was observed in keratinocytes after 'IV08.009' application. CONCLUSION Basal keratinocytes may divide frequently during skin lifespan, and signs of deterioration could appear such as loss of protein factors required for correct mitosis. Our findings suggest that mitotic abnormalities can be prevented by the modulation of CRM1 and survivin. We demonstrated the ability of compound 'IV08.009' to efficiently protect cultured keratinocytes from mitotic abnormalities.
Collapse
Affiliation(s)
- F Labarrade
- Ashland Specialty Ingredients, Vincience Global Skin Research Center, 655, route du Pin Montard, 06904 Sophia Antipolis, France
| | - J-M Botto
- Ashland Specialty Ingredients, Vincience Global Skin Research Center, 655, route du Pin Montard, 06904 Sophia Antipolis, France.
| | - N Domloge
- Ashland Specialty Ingredients, Vincience Global Skin Research Center, 655, route du Pin Montard, 06904 Sophia Antipolis, France
| |
Collapse
|
32
|
Wu Z, Jin Z, Zhang X, Shen N, Wang J, Zhao Y, Mei L. Nup62, associated with spindle microtubule rather than spindle matrix, is involved in chromosome alignment and spindle assembly during mitosis. Cell Biol Int 2016; 40:968-75. [PMID: 27298184 DOI: 10.1002/cbin.10633] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Accepted: 06/01/2016] [Indexed: 11/10/2022]
Abstract
An increasing number of the active mitotic functions of nucleoporins in the distinct steps of mitosis have been assigned over the past few years. As one of FG-repeats containing nucleoporins, Nup62 has been found to be involved in nuclear transport, cell migration, virus infection, and cell cycle regulation. However, the role and mechanism of Nup62 in mitotic regulation have not been fully revealed. In this paper, it was revealed that a fraction of Nup62 was associated with mitotic spindle microtubule instead of spindle matrix, and the localization of Nup62 in the mitotic spindle depended on its three coiled-coil domains rather than Crm1, although Nup62 strongly interacted with Crm1 during mitosis. Moreover, depletion of Nup62 by small interference of RNA seriously induced the defects of chromosome alignment and spindle assembly although the bipolar spindle was observed in most of the Nup62 knock-down cells. Notably, congression of polar chromosome defect was observed in more than 30% of Nup62 knock-down cells. These findings revealed that Nup62 was a novel mitotic spindle associated nucleoporin and involved in chromosome alignment and spindle assembly.
Collapse
Affiliation(s)
- Zhige Wu
- Department of Biotechnology and Chemical Engineering, Ningbo Institute of Technology, Zhejiang University, Ningbo 315100, China
| | - Zhihua Jin
- Department of Biotechnology and Chemical Engineering, Ningbo Institute of Technology, Zhejiang University, Ningbo 315100, China
| | - Xinhong Zhang
- Department of Biotechnology and Chemical Engineering, Ningbo Institute of Technology, Zhejiang University, Ningbo 315100, China
| | - Na Shen
- Department of Biotechnology and Chemical Engineering, Ningbo Institute of Technology, Zhejiang University, Ningbo 315100, China
| | - Jinbo Wang
- Department of Biotechnology and Chemical Engineering, Ningbo Institute of Technology, Zhejiang University, Ningbo 315100, China
| | - Yingxian Zhao
- Department of Biotechnology and Chemical Engineering, Ningbo Institute of Technology, Zhejiang University, Ningbo 315100, China
| | - Lehe Mei
- Department of Biotechnology and Chemical Engineering, Ningbo Institute of Technology, Zhejiang University, Ningbo 315100, China
| |
Collapse
|
33
|
A Vulnerability of a Subset of Colon Cancers with Potential Clinical Utility. Cell 2016; 165:317-30. [PMID: 27058664 DOI: 10.1016/j.cell.2016.02.059] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2015] [Revised: 12/20/2015] [Accepted: 02/22/2016] [Indexed: 01/11/2023]
Abstract
BRAF(V600E) mutant colon cancers (CCs) have a characteristic gene expression signature that is also found in some tumors lacking this mutation. Collectively, they are referred to as "BRAF-like" tumors and represent some 20% of CCs. We used a shRNA-based genetic screen focused on genes upregulated in BRAF(V600E) CCs to identify vulnerabilities of this tumor subtype that might be exploited therapeutically. Here, we identify RANBP2 (also known as NUP358) as essential for survival of BRAF-like, but not for non-BRAF-like, CC cells. Suppression of RANBP2 results in mitotic defects only in BRAF-like CC cells, leading to cell death. Mechanistically, RANBP2 silencing reduces microtubule outgrowth from the kinetochores, thereby inducing spindle perturbations, providing an explanation for the observed mitotic defects. We find that BRAF-like CCs display far greater sensitivity to the microtubule poison vinorelbine both in vitro and in vivo, suggesting that vinorelbine is a potential tailored treatment for BRAF-like CCs.
Collapse
|
34
|
Ritterhoff T, Das H, Hofhaus G, Schröder RR, Flotho A, Melchior F. The RanBP2/RanGAP1*SUMO1/Ubc9 SUMO E3 ligase is a disassembly machine for Crm1-dependent nuclear export complexes. Nat Commun 2016; 7:11482. [PMID: 27160050 PMCID: PMC4866044 DOI: 10.1038/ncomms11482] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2015] [Accepted: 03/31/2016] [Indexed: 02/07/2023] Open
Abstract
Continuous cycles of nucleocytoplasmic transport require disassembly of transport receptor/Ran-GTP complexes in the cytoplasm. A basic disassembly mechanism in all eukaryotes depends on soluble RanGAP and RanBP1. In vertebrates, a significant fraction of RanGAP1 stably interacts with the nucleoporin RanBP2 at a binding site that is flanked by FG-repeats and Ran-binding domains, and overlaps with RanBP2's SUMO E3 ligase region. Here, we show that the RanBP2/RanGAP1*SUMO1/Ubc9 complex functions as an autonomous disassembly machine with a preference for the export receptor Crm1. We describe three in vitro reconstituted disassembly intermediates, which show binding of a Crm1 export complex via two FG-repeat patches, cargo-release by RanBP2's Ran-binding domains and retention of free Crm1 at RanBP2 after Ran-GTP hydrolysis. Intriguingly, all intermediates are compatible with SUMO E3 ligase activity, suggesting that the RanBP2/RanGAP1*SUMO1/Ubc9 complex may link Crm1- and SUMO-dependent functions.
Collapse
Affiliation(s)
- Tobias Ritterhoff
- Zentrum für Molekulare Biologie der Universität Heidelberg, DKFZ-ZMBH Alliance, Heidelberg 69120, Germany
- Department of Biochemistry, University of Washington, Seattle, Washington 98195, USA
| | - Hrishikesh Das
- Zentrum für Molekulare Biologie der Universität Heidelberg, DKFZ-ZMBH Alliance, Heidelberg 69120, Germany
- Cryo Electron Microscopy, CellNetworks, BioQuant, Universitätsklinikum Heidelberg, Heidelberg 69120, Germany
| | - Götz Hofhaus
- Cryo Electron Microscopy, CellNetworks, BioQuant, Universitätsklinikum Heidelberg, Heidelberg 69120, Germany
| | - Rasmus R. Schröder
- Cryo Electron Microscopy, CellNetworks, BioQuant, Universitätsklinikum Heidelberg, Heidelberg 69120, Germany
| | - Annette Flotho
- Zentrum für Molekulare Biologie der Universität Heidelberg, DKFZ-ZMBH Alliance, Heidelberg 69120, Germany
| | - Frauke Melchior
- Zentrum für Molekulare Biologie der Universität Heidelberg, DKFZ-ZMBH Alliance, Heidelberg 69120, Germany
| |
Collapse
|
35
|
HIV-1 and M-PMV RNA Nuclear Export Elements Program Viral Genomes for Distinct Cytoplasmic Trafficking Behaviors. PLoS Pathog 2016; 12:e1005565. [PMID: 27070420 PMCID: PMC4829213 DOI: 10.1371/journal.ppat.1005565] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2015] [Accepted: 03/21/2016] [Indexed: 12/15/2022] Open
Abstract
Retroviruses encode cis-acting RNA nuclear export elements that override nuclear retention of intron-containing viral mRNAs including the full-length, unspliced genomic RNAs (gRNAs) packaged into assembling virions. The HIV-1 Rev-response element (RRE) recruits the cellular nuclear export receptor CRM1 (also known as exportin-1/XPO1) using the viral protein Rev, while simple retroviruses encode constitutive transport elements (CTEs) that directly recruit components of the NXF1(Tap)/NXT1(p15) mRNA nuclear export machinery. How gRNA nuclear export is linked to trafficking machineries in the cytoplasm upstream of virus particle assembly is unknown. Here we used long-term (>24 h), multicolor live cell imaging to directly visualize HIV-1 gRNA nuclear export, translation, cytoplasmic trafficking, and virus particle production in single cells. We show that the HIV-1 RRE regulates unique, en masse, Rev- and CRM1-dependent "burst-like" transitions of mRNAs from the nucleus to flood the cytoplasm in a non-localized fashion. By contrast, the CTE derived from Mason-Pfizer monkey virus (M-PMV) links gRNAs to microtubules in the cytoplasm, driving them to cluster markedly to the centrosome that forms the pericentriolar core of the microtubule-organizing center (MTOC). Adding each export element to selected heterologous mRNAs was sufficient to confer each distinct export behavior, as was directing Rev/CRM1 or NXF1/NXT1 transport modules to mRNAs using a site-specific RNA tethering strategy. Moreover, multiple CTEs per transcript enhanced MTOC targeting, suggesting that a cooperative mechanism links NXF1/NXT1 to microtubules. Combined, these results reveal striking, unexpected features of retroviral gRNA nucleocytoplasmic transport and demonstrate roles for mRNA export elements that extend beyond nuclear pores to impact gRNA distribution in the cytoplasm.
Collapse
|
36
|
Nakayama R, Zhang YX, Czaplinski JT, Anatone AJ, Sicinska ET, Fletcher JA, Demetri GD, Wagner AJ. Preclinical activity of selinexor, an inhibitor of XPO1, in sarcoma. Oncotarget 2016; 7:16581-92. [PMID: 26918731 PMCID: PMC4941336 DOI: 10.18632/oncotarget.7667] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2015] [Accepted: 02/09/2016] [Indexed: 12/11/2022] Open
Abstract
Selinexor is an orally bioavailable selective inhibitor of nuclear export that has been demonstrated to have preclinical activity in various cancer types and that is currently in Phase I and II clinical trials for advanced cancers. In this study, we evaluated the effects of selinexor in several preclinical models of various sarcoma subtypes. The efficacy of selinexor was investigated in vitro and in vivo using 17 cell lines and 9 sarcoma xenograft models including gastrointestinal stromal tumor (GIST), liposarcoma (LPS), leiomyosarcoma, rhabdomyosarcoma, undifferentiated sarcomas, and alveolar soft part sarcoma (ASPS). Most sarcoma cell lines were sensitive to selinexor with IC50s ranging from 28.8 nM to 218.2 nM (median: 66.1 nM). Selinexor suppressed sarcoma tumor xenograft growth, including models of ASPS that were resistant in vitro. In GIST cells with KIT mutations, selinexor induced G1- arrest without attenuation of phosphorylation of KIT, AKT, or MAPK, in contrast to imatinib. In LPS cell lines with MDM2 and CDK4 amplification, selinexor induced G1-arrest and apoptosis irrespective of p53 expression or mutation and irrespective of RB expression. Selinexor increased p53 and p21 expression at the protein but not RNA level, indicating a post-transcriptional effect. These results indicate that selinexor has potent in vitro and in vivo activity against a wide variety of sarcoma models by inducing G1-arrest independent of known molecular mechanisms in GIST and LPS. These studies further justify the exploration of selinexor in clinical trials targeting various sarcoma subtypes.
Collapse
Affiliation(s)
- Robert Nakayama
- Ludwig Center at Dana-Farber/Harvard and Center for Sarcoma and Bone Oncology, Department of Medical Oncology, Harvard Medical School, Boston, MA, USA
- Department of Orthopaedic Surgery, School of Medicine, Keio University, Tokyo, Japan
| | - Yi-Xiang Zhang
- Ludwig Center at Dana-Farber/Harvard and Center for Sarcoma and Bone Oncology, Department of Medical Oncology, Harvard Medical School, Boston, MA, USA
| | - Jeffrey T. Czaplinski
- Department of Medical Oncology and Center for Molecular Oncologic Pathology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Alex J. Anatone
- Department of Medical Oncology and Center for Molecular Oncologic Pathology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Ewa T. Sicinska
- Department of Medical Oncology and Center for Molecular Oncologic Pathology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Jonathan A. Fletcher
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - George D. Demetri
- Ludwig Center at Dana-Farber/Harvard and Center for Sarcoma and Bone Oncology, Department of Medical Oncology, Harvard Medical School, Boston, MA, USA
| | - Andrew J. Wagner
- Ludwig Center at Dana-Farber/Harvard and Center for Sarcoma and Bone Oncology, Department of Medical Oncology, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
37
|
Vaidyanathan S, Thangavelu PU, Duijf PHG. Overexpression of Ran GTPase Components Regulating Nuclear Export, but not Mitotic Spindle Assembly, Marks Chromosome Instability and Poor Prognosis in Breast Cancer. Target Oncol 2016; 11:677-686. [DOI: 10.1007/s11523-016-0432-y] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
38
|
Fernandes JD, Booth DS, Frankel AD. A structurally plastic ribonucleoprotein complex mediates post-transcriptional gene regulation in HIV-1. WILEY INTERDISCIPLINARY REVIEWS-RNA 2016; 7:470-86. [PMID: 26929078 DOI: 10.1002/wrna.1342] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Revised: 01/12/2016] [Accepted: 01/14/2016] [Indexed: 01/28/2023]
Abstract
HIV replication requires the nuclear export of essential, intron-containing viral RNAs. To facilitate export, HIV encodes the viral accessory protein Rev which binds unspliced and partially spliced viral RNAs and creates a ribonucleoprotein complex that recruits the cellular Chromosome maintenance factor 1 export machinery. Exporting RNAs in this manner bypasses the necessity for complete splicing as a prerequisite for mRNA export, and allows intron-containing RNAs to reach the cytoplasm intact for translation and virus packaging. Recent structural studies have revealed that this entire complex exhibits remarkable plasticity at many levels of organization, including RNA folding, protein-RNA recognition, multimer formation, and host factor recruitment. In this review, we explore each aspect of plasticity from structural, functional, and possible therapeutic viewpoints. WIREs RNA 2016, 7:470-486. doi: 10.1002/wrna.1342 For further resources related to this article, please visit the WIREs website.
Collapse
Affiliation(s)
- Jason D Fernandes
- Department of Biochemistry and Biophysics, University of California, San Francisco, CA, USA
| | - David S Booth
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA
| | - Alan D Frankel
- Department of Biochemistry and Biophysics, University of California, San Francisco, CA, USA
| |
Collapse
|
39
|
Cairo L, Wozniak R. The Nuclear Transport Factor Kap121 Is Required for Stability of the Dam1 Complex and Mitotic Kinetochore Bi-orientation. Cell Rep 2016; 14:2440-50. [DOI: 10.1016/j.celrep.2016.02.041] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2015] [Revised: 11/04/2015] [Accepted: 02/04/2016] [Indexed: 12/30/2022] Open
|
40
|
Cavazza T, Vernos I. The RanGTP Pathway: From Nucleo-Cytoplasmic Transport to Spindle Assembly and Beyond. Front Cell Dev Biol 2016; 3:82. [PMID: 26793706 PMCID: PMC4707252 DOI: 10.3389/fcell.2015.00082] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Accepted: 12/07/2015] [Indexed: 01/03/2023] Open
Abstract
The small GTPase Ran regulates the interaction of transport receptors with a number of cellular cargo proteins. The high affinity binding of the GTP-bound form of Ran to import receptors promotes cargo release, whereas its binding to export receptors stabilizes their interaction with the cargo. This basic mechanism linked to the asymmetric distribution of the two nucleotide-bound forms of Ran between the nucleus and the cytoplasm generates a switch like mechanism controlling nucleo-cytoplasmic transport. Since 1999, we have known that after nuclear envelope breakdown (NEBD) Ran and the above transport receptors also provide a local control over the activity of factors driving spindle assembly and regulating other aspects of cell division. The identification and functional characterization of RanGTP mitotic targets is providing novel insights into mechanisms essential for cell division. Here we review our current knowledge on the RanGTP system and its regulation and we focus on the recent advances made through the characterization of its mitotic targets. We then briefly review the novel functions of the pathway that were recently described. Altogether, the RanGTP system has moonlighting functions exerting a spatial control over protein interactions that drive specific functions depending on the cellular context.
Collapse
Affiliation(s)
- Tommaso Cavazza
- Cell and Developmental Biology, Centre for Genomic Regulation, The Barcelona Institute of Science and TechnologyBarcelona, Spain; Universitat Pompeu FabraBarcelona, Spain
| | - Isabelle Vernos
- Cell and Developmental Biology, Centre for Genomic Regulation, The Barcelona Institute of Science and TechnologyBarcelona, Spain; Universitat Pompeu FabraBarcelona, Spain; Institució Catalana de Recerca I Estudis AvançatsBarcelona, Spain
| |
Collapse
|
41
|
Kırlı K, Karaca S, Dehne HJ, Samwer M, Pan KT, Lenz C, Urlaub H, Görlich D. A deep proteomics perspective on CRM1-mediated nuclear export and nucleocytoplasmic partitioning. eLife 2015; 4:e11466. [PMID: 26673895 PMCID: PMC4764573 DOI: 10.7554/elife.11466] [Citation(s) in RCA: 161] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Accepted: 12/13/2015] [Indexed: 12/23/2022] Open
Abstract
CRM1 is a highly conserved, RanGTPase-driven exportin that carries proteins and RNPs from the nucleus to the cytoplasm. We now explored the cargo-spectrum of CRM1 in depth and identified surprisingly large numbers, namely >700 export substrates from the yeast S. cerevisiae, ≈1000 from Xenopus oocytes and >1050 from human cells. In addition, we quantified the partitioning of ≈5000 unique proteins between nucleus and cytoplasm of Xenopus oocytes. The data suggest new CRM1 functions in spatial control of vesicle coat-assembly, centrosomes, autophagy, peroxisome biogenesis, cytoskeleton, ribosome maturation, translation, mRNA degradation, and more generally in precluding a potentially detrimental action of cytoplasmic pathways within the nuclear interior. There are also numerous new instances where CRM1 appears to act in regulatory circuits. Altogether, our dataset allows unprecedented insights into the nucleocytoplasmic organisation of eukaryotic cells, into the contributions of an exceedingly promiscuous exportin and it provides a new basis for NES prediction.
Collapse
Affiliation(s)
- Koray Kırlı
- Department of Cellular Logistics, Max Planck Institute for Biophysical Chemistry, Göttingen, Germany
| | - Samir Karaca
- Department of Cellular Logistics, Max Planck Institute for Biophysical Chemistry, Göttingen, Germany
- Bioanalytical Mass Spectrometry Group, Max Planck Institute for Biophysical Chemistry, Göttingen, Germany
| | - Heinz Jürgen Dehne
- Department of Cellular Logistics, Max Planck Institute for Biophysical Chemistry, Göttingen, Germany
| | - Matthias Samwer
- Department of Cellular Logistics, Max Planck Institute for Biophysical Chemistry, Göttingen, Germany
| | - Kuan Ting Pan
- Bioanalytical Mass Spectrometry Group, Max Planck Institute for Biophysical Chemistry, Göttingen, Germany
| | - Christof Lenz
- Bioanalytical Mass Spectrometry Group, Max Planck Institute for Biophysical Chemistry, Göttingen, Germany
- Bioanalytics, Institute for Clinical Chemistry, University Medical Center Göttingen, Göttingen, Germany
| | - Henning Urlaub
- Bioanalytical Mass Spectrometry Group, Max Planck Institute for Biophysical Chemistry, Göttingen, Germany
- Bioanalytics, Institute for Clinical Chemistry, University Medical Center Göttingen, Göttingen, Germany
| | - Dirk Görlich
- Department of Cellular Logistics, Max Planck Institute for Biophysical Chemistry, Göttingen, Germany
| |
Collapse
|
42
|
Complex Commingling: Nucleoporins and the Spindle Assembly Checkpoint. Cells 2015; 4:706-25. [PMID: 26540075 PMCID: PMC4695854 DOI: 10.3390/cells4040706] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Revised: 10/12/2015] [Accepted: 10/28/2015] [Indexed: 12/14/2022] Open
Abstract
The segregation of the chromosomes during mitosis is an important process, in which the replicated DNA content is properly allocated into two daughter cells. To ensure their genomic integrity, cells present an essential surveillance mechanism known as the spindle assembly checkpoint (SAC), which monitors the bipolar attachment of the mitotic spindle to chromosomes to prevent errors that would result in chromosome mis-segregation and aneuploidy. Multiple components of the nuclear pore complex (NPC), a gigantic protein complex that forms a channel through the nuclear envelope to allow nucleocytoplasmic exchange of macromolecules, were shown to be critical for faithful cell division and implicated in the regulation of different steps of the mitotic process, including kinetochore and spindle assembly as well as the SAC. In this review, we will describe current knowledge about the interconnection between the NPC and the SAC in an evolutional perspective, which primarily relies on the two mitotic checkpoint regulators, Mad1 and Mad2. We will further discuss the role of NPC constituents, the nucleoporins, in kinetochore and spindle assembly and the formation of the mitotic checkpoint complex during mitosis and interphase.
Collapse
|
43
|
Dickmanns A, Monecke T, Ficner R. Structural Basis of Targeting the Exportin CRM1 in Cancer. Cells 2015; 4:538-68. [PMID: 26402707 PMCID: PMC4588050 DOI: 10.3390/cells4030538] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Revised: 09/07/2015] [Accepted: 09/11/2015] [Indexed: 12/19/2022] Open
Abstract
Recent studies have demonstrated the interference of nucleocytoplasmic trafficking with the establishment and maintenance of various cancers. Nucleocytoplasmic transport is highly regulated and coordinated, involving different nuclear transport factors or receptors, importins and exportins, that mediate cargo transport from the cytoplasm into the nucleus or the other way round, respectively. The exportin CRM1 (Chromosome region maintenance 1) exports a plethora of different protein cargoes and ribonucleoprotein complexes. Structural and biochemical analyses have enabled the deduction of individual steps of the CRM1 transport cycle. In addition, CRM1 turned out to be a valid target for anticancer drugs as it exports numerous proto-oncoproteins and tumor suppressors. Clearly, detailed understanding of the flexibility, regulatory features and cooperative binding properties of CRM1 for Ran and cargo is a prerequisite for the design of highly effective drugs. The first compound found to inhibit CRM1-dependent nuclear export was the natural drug Leptomycin B (LMB), which blocks export by competitively interacting with a highly conserved cleft on CRM1 required for nuclear export signal recognition. Clinical studies revealed serious side effects of LMB, leading to a search for alternative natural and synthetic drugs and hence a multitude of novel therapeutics. The present review examines recent progress in understanding the binding mode of natural and synthetic compounds and their inhibitory effects.
Collapse
Affiliation(s)
- Achim Dickmanns
- Abteilung für Molekulare Strukturbiologie, Institut für Mikrobiologie und Genetik, GZMB, Georg-August-Universität Göttingen, Justus-von-Liebig-Weg 11, Göttingen 37077, Germany.
| | - Thomas Monecke
- Abteilung für Molekulare Strukturbiologie, Institut für Mikrobiologie und Genetik, GZMB, Georg-August-Universität Göttingen, Justus-von-Liebig-Weg 11, Göttingen 37077, Germany.
| | - Ralf Ficner
- Abteilung für Molekulare Strukturbiologie, Institut für Mikrobiologie und Genetik, GZMB, Georg-August-Universität Göttingen, Justus-von-Liebig-Weg 11, Göttingen 37077, Germany.
| |
Collapse
|
44
|
Forbes DJ, Travesa A, Nord MS, Bernis C. Reprint of "Nuclear transport factors: global regulation of mitosis". Curr Opin Cell Biol 2015. [PMID: 26196321 DOI: 10.1016/j.ceb.2015.07.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
The unexpected repurposing of nuclear transport proteins from their function in interphase to an equally vital and very different set of functions in mitosis was very surprising. The multi-talented cast when first revealed included the import receptors, importin alpha and beta, the small regulatory GTPase RanGTP, and a subset of nuclear pore proteins. In this review, we report that recent years have revealed new discoveries in each area of this expanding story in vertebrates: (a) The cast of nuclear import receptors playing a role in mitotic spindle regulation has expanded: both transportin, a nuclear import receptor, and Crm1/Xpo1, an export receptor, are involved in different aspects of spindle assembly. Importin beta and transportin also regulate nuclear envelope and pore assembly. (b) The role of nucleoporins has grown to include recruiting the key microtubule nucleator – the γ-TuRC complex – and the exportin Crm1 to the mitotic kinetochores of humans. Together they nucleate microtubule formation from the kinetochores toward the centrosomes. (c) New research finds that the original importin beta/RanGTP team have been further co-opted by evolution to help regulate other cellular and organismal activities, ranging from the actual positioning of the spindle within the cell perimeter, to regulation of a newly discovered spindle microtubule branching activity, to regulation of the interaction of microtubule structures with specific actin structures. (d) Lastly, because of the multitudinous roles of karyopherins throughout the cell cycle, a recent large push toward testing their potential as chemotherapeutic targets has begun to yield burgeoning progress in the clinic.
Collapse
Affiliation(s)
- Douglass J Forbes
- Section of Cell and Developmental Biology, Division of Biological Sciences 0347, Room 2124A Pacific Hall, University of California-San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0347, United States.
| | - Anna Travesa
- Section of Cell and Developmental Biology, Division of Biological Sciences 0347, Room 2124A Pacific Hall, University of California-San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0347, United States
| | - Matthew S Nord
- Section of Cell and Developmental Biology, Division of Biological Sciences 0347, Room 2124A Pacific Hall, University of California-San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0347, United States
| | - Cyril Bernis
- Section of Cell and Developmental Biology, Division of Biological Sciences 0347, Room 2124A Pacific Hall, University of California-San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0347, United States
| |
Collapse
|
45
|
Forbes DJ, Travesa A, Nord MS, Bernis C. Nuclear transport factors: global regulation of mitosis. Curr Opin Cell Biol 2015; 35:78-90. [PMID: 25982429 DOI: 10.1016/j.ceb.2015.04.012] [Citation(s) in RCA: 85] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2015] [Revised: 04/01/2015] [Accepted: 04/17/2015] [Indexed: 12/22/2022]
Abstract
The unexpected repurposing of nuclear transport proteins from their function in interphase to an equally vital and very different set of functions in mitosis was very surprising. The multi-talented cast when first revealed included the import receptors, importin alpha and beta, the small regulatory GTPase RanGTP, and a subset of nuclear pore proteins. In this review, we report that recent years have revealed new discoveries in each area of this expanding story in vertebrates: (a) The cast of nuclear import receptors playing a role in mitotic spindle regulation has expanded: both transportin, a nuclear import receptor, and Crm1/Xpo1, an export receptor, are involved in different aspects of spindle assembly. Importin beta and transportin also regulate nuclear envelope and pore assembly. (b) The role of nucleoporins has grown to include recruiting the key microtubule nucleator - the γ-TuRC complex - and the exportin Crm1 to the mitotic kinetochores of humans. Together they nucleate microtubule formation from the kinetochores toward the centrosomes. (c) New research finds that the original importin beta/RanGTP team have been further co-opted by evolution to help regulate other cellular and organismal activities, ranging from the actual positioning of the spindle within the cell perimeter, to regulation of a newly discovered spindle microtubule branching activity, to regulation of the interaction of microtubule structures with specific actin structures. (d) Lastly, because of the multitudinous roles of karyopherins throughout the cell cycle, a recent large push toward testing their potential as chemotherapeutic targets has begun to yield burgeoning progress in the clinic.
Collapse
Affiliation(s)
- Douglass J Forbes
- Section of Cell and Developmental Biology, Division of Biological Sciences 0347, Room 2124A Pacific Hall, University of California-San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0347, United States.
| | - Anna Travesa
- Section of Cell and Developmental Biology, Division of Biological Sciences 0347, Room 2124A Pacific Hall, University of California-San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0347, United States
| | - Matthew S Nord
- Section of Cell and Developmental Biology, Division of Biological Sciences 0347, Room 2124A Pacific Hall, University of California-San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0347, United States
| | - Cyril Bernis
- Section of Cell and Developmental Biology, Division of Biological Sciences 0347, Room 2124A Pacific Hall, University of California-San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0347, United States
| |
Collapse
|
46
|
Khorashad JS, Eiring AM, Mason CC, Gantz KC, Bowler AD, Redwine HM, Yu F, Kraft IL, Pomicter AD, Reynolds KR, Iovino AJ, Zabriskie MS, Heaton WL, Tantravahi SK, Kauffman M, Shacham S, Chenchik A, Bonneau K, Ullman KS, O'Hare T, Deininger MW. shRNA library screening identifies nucleocytoplasmic transport as a mediator of BCR-ABL1 kinase-independent resistance. Blood 2015; 125:1772-81. [PMID: 25573989 PMCID: PMC4357584 DOI: 10.1182/blood-2014-08-588855] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2014] [Accepted: 12/23/2014] [Indexed: 12/26/2022] Open
Abstract
The mechanisms underlying tyrosine kinase inhibitor (TKI) resistance in chronic myeloid leukemia (CML) patients lacking explanatory BCR-ABL1 kinase domain mutations are incompletely understood. To identify mechanisms of TKI resistance that are independent of BCR-ABL1 kinase activity, we introduced a lentiviral short hairpin RNA (shRNA) library targeting ∼5000 cell signaling genes into K562(R), a CML cell line with BCR-ABL1 kinase-independent TKI resistance expressing exclusively native BCR-ABL1. A customized algorithm identified genes whose shRNA-mediated knockdown markedly impaired growth of K562(R) cells compared with TKI-sensitive controls. Among the top candidates were 2 components of the nucleocytoplasmic transport complex, RAN and XPO1 (CRM1). shRNA-mediated RAN inhibition or treatment of cells with the XPO1 inhibitor, KPT-330 (Selinexor), increased the imatinib sensitivity of CML cell lines with kinase-independent TKI resistance. Inhibition of either RAN or XPO1 impaired colony formation of CD34(+) cells from newly diagnosed and TKI-resistant CML patients in the presence of imatinib, without effects on CD34(+) cells from normal cord blood or from a patient harboring the BCR-ABL1(T315I) mutant. These data implicate RAN in BCR-ABL1 kinase-independent imatinib resistance and show that shRNA library screens are useful to identify alternative pathways critical to drug resistance in CML.
Collapse
MESH Headings
- Active Transport, Cell Nucleus/genetics
- Benzamides/pharmacology
- Cell Line, Tumor
- Cell Survival
- Drug Resistance, Neoplasm/genetics
- Fusion Proteins, bcr-abl/genetics
- Fusion Proteins, bcr-abl/metabolism
- Gene Knockdown Techniques
- Gene Library
- Humans
- Hydrazines/pharmacology
- Imatinib Mesylate
- K562 Cells
- Karyopherins/antagonists & inhibitors
- Karyopherins/genetics
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism
- Mutation
- Piperazines/pharmacology
- Protein Kinase Inhibitors/pharmacology
- Pyrimidines/pharmacology
- RNA, Small Interfering/genetics
- Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors
- Receptors, Cytoplasmic and Nuclear/genetics
- Signal Transduction
- Triazoles/pharmacology
- Tumor Stem Cell Assay
- ran GTP-Binding Protein/antagonists & inhibitors
- ran GTP-Binding Protein/genetics
- Exportin 1 Protein
Collapse
Affiliation(s)
| | - Anna M Eiring
- Huntsman Cancer Institute, The University of Utah, Salt Lake City, UT
| | - Clinton C Mason
- Huntsman Cancer Institute, The University of Utah, Salt Lake City, UT
| | - Kevin C Gantz
- Huntsman Cancer Institute, The University of Utah, Salt Lake City, UT
| | - Amber D Bowler
- Huntsman Cancer Institute, The University of Utah, Salt Lake City, UT
| | - Hannah M Redwine
- Huntsman Cancer Institute, The University of Utah, Salt Lake City, UT
| | - Fan Yu
- Huntsman Cancer Institute, The University of Utah, Salt Lake City, UT; Beijing Tsinghua Chang Gung Hospital, Tsinghua University, Beijing, China
| | - Ira L Kraft
- Huntsman Cancer Institute, The University of Utah, Salt Lake City, UT
| | | | | | - Anthony J Iovino
- Huntsman Cancer Institute, The University of Utah, Salt Lake City, UT
| | | | - William L Heaton
- Huntsman Cancer Institute, The University of Utah, Salt Lake City, UT
| | - Srinivas K Tantravahi
- Huntsman Cancer Institute, The University of Utah, Salt Lake City, UT; Division of Hematology and Hematologic Malignancies, The University of Utah, Salt Lake City, UT
| | | | | | | | | | | | - Thomas O'Hare
- Huntsman Cancer Institute, The University of Utah, Salt Lake City, UT; Division of Hematology and Hematologic Malignancies, The University of Utah, Salt Lake City, UT
| | - Michael W Deininger
- Huntsman Cancer Institute, The University of Utah, Salt Lake City, UT; Division of Hematology and Hematologic Malignancies, The University of Utah, Salt Lake City, UT
| |
Collapse
|
47
|
Jia H, Zhang X, Wang W, Bai Y, Ling Y, Cao C, Ma RZ, Zhong H, Wang X, Xu Q. A putative N-terminal nuclear export sequence is sufficient for Mps1 nuclear exclusion during interphase. BMC Cell Biol 2015; 16:6. [PMID: 25886724 PMCID: PMC4373099 DOI: 10.1186/s12860-015-0048-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2014] [Accepted: 01/13/2015] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Mps1, an essential component of the mitotic checkpoint, is also an important interphase regulator and has roles in DNA damage response, cytokinesis and centrosome duplication. Mps1 predominantly resides in the cytoplasm and relocates into the nucleus at the late G2 phase. So far, the mechanism underlying the Mps1 translocation between the cytoplasm and nucleus has been unclear. RESULTS In this work, a dynamic export process of Mps1 from the nucleus to cytoplasm in interphase was revealed- a process blocked by the Crm1 inhibitor, Leptomycin B, suggesting that export of Mps1 is Crm1 dependent. Consistent with this speculation, a direct association between Mps1 and Crm1 was found. Furthermore, a putative nuclear export sequence (pNES) motif at the N-terminal of Mps1 was identified by analyzing the motif of Mps1. This motif shows a high sequence similarity to the classic NES, a fusion of this motif with EGFP results in dramatic exclusion of the fusion protein from the nucleus. Additionally, Mps1 mutant loss of pNES integrity was shown by replacing leucine with alanine which produced a diffused subcellular distribution, compared to the wild type protein which resides predominantly in cytoplasm. CONCLUSION Taken these findings together, it was concluded that the pNES sequence is sufficient for the Mps1 export from nucleus during interphase.
Collapse
Affiliation(s)
- Haiwei Jia
- Navy General Hospital of China, Beijing, 100048, China.
| | - Xiaojuan Zhang
- State Key Laboratory for Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China.
| | - Wenjun Wang
- Beijing Institute of Biotechnology, Taiping road 27, POB 130(8), Beijing, 100850, China.
| | - Yuanyuan Bai
- Beijing Institute of Biotechnology, Taiping road 27, POB 130(8), Beijing, 100850, China.
| | - Youguo Ling
- Beijing Institute of Biotechnology, Taiping road 27, POB 130(8), Beijing, 100850, China.
- Department of Life Science, Anhui University, Hefei, 230601, China.
| | - Cheng Cao
- Beijing Institute of Biotechnology, Taiping road 27, POB 130(8), Beijing, 100850, China.
| | - Runlin Z Ma
- State Key Laboratory for Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China.
| | - Hui Zhong
- Beijing Institute of Biotechnology, Taiping road 27, POB 130(8), Beijing, 100850, China.
| | - Xue Wang
- West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Quanbin Xu
- Beijing Institute of Biotechnology, Taiping road 27, POB 130(8), Beijing, 100850, China.
| |
Collapse
|
48
|
Markossian S, Suresh S, Osmani AH, Osmani SA. Nup2 requires a highly divergent partner, NupA, to fulfill functions at nuclear pore complexes and the mitotic chromatin region. Mol Biol Cell 2014; 26:605-21. [PMID: 25540430 PMCID: PMC4325833 DOI: 10.1091/mbc.e14-09-1359] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Among nuclear pore proteins, Nup2 is unique because it transfers to the mitotic chromatin region to fulfill unknown functions. Analysis of Nup2 and a novel targeting partner, NupA, shows that they are required for normal anaphase and nucleokinesis. Their functions also involve an import pathway for Mad1 but apparently not general nuclear protein import. Chromatin and nuclear pore complexes (NPCs) undergo dramatic changes during mitosis, which in vertebrates and Aspergillus nidulans involves movement of Nup2 from NPCs to the chromatin region to fulfill unknown functions. This transition is shown to require the Cdk1 mitotic kinase and be promoted prematurely by ectopic expression of the NIMA kinase. Nup2 localizes with a copurifying partner termed NupA, a highly divergent yet essential NPC protein. NupA and Nup2 locate throughout the chromatin region during prophase but during anaphase move to surround segregating DNA. NupA function is shown to involve targeting Nup2 to its interphase and mitotic locations. Deletion of either Nup2 or NupA causes identical mitotic defects that initiate a spindle assembly checkpoint (SAC)–dependent mitotic delay and also cause defects in karyokinesis. These mitotic problems are not caused by overall defects in mitotic NPC disassembly–reassembly or general nuclear import. However, without Nup2 or NupA, although the SAC protein Mad1 locates to its mitotic locations, it fails to locate to NPCs normally in G1 after mitosis. Collectively the study provides new insight into the roles of Nup2 and NupA during mitosis and in a surveillance mechanism that regulates nucleokinesis when mitotic defects occur after SAC fulfillment.
Collapse
Affiliation(s)
- Sarine Markossian
- Laboratory of Gene Regulation and Development, National Institute for Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892
| | | | - Aysha H Osmani
- Department of Molecular Genetics, Ohio State University, Columbus, OH 43210
| | - Stephen A Osmani
- Department of Molecular Genetics, Ohio State University, Columbus, OH 43210
| |
Collapse
|
49
|
Cautain B, Hill R, de Pedro N, Link W. Components and regulation of nuclear transport processes. FEBS J 2014; 282:445-62. [PMID: 25429850 PMCID: PMC7163960 DOI: 10.1111/febs.13163] [Citation(s) in RCA: 177] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Revised: 11/11/2014] [Accepted: 11/12/2014] [Indexed: 12/27/2022]
Abstract
The spatial separation of DNA replication and gene transcription in the nucleus and protein translation in the cytoplasm is a uniform principle of eukaryotic cells. This compartmentalization imposes a requirement for a transport network of macromolecules to shuttle these components in and out of the nucleus. This nucleo‐cytoplasmic transport of macromolecules is critical for both cell physiology and pathology. Consequently, investigating its regulation and disease‐associated alterations can reveal novel therapeutic approaches to fight human diseases, such as cancer or viral infection. The characterization of the nuclear pore complex, the identification of transport signals and transport receptors, as well as the characterization of the Ran system (providing the energy source for efficient cargo transport) has greatly facilitated our understanding of the components, mechanisms and regulation of the nucleo‐cytoplasmic transport of proteins in our cells. Here we review this knowledge with a specific emphasis on the selection of disease‐relevant molecular targets for potential therapeutic intervention.
Collapse
Affiliation(s)
- Bastien Cautain
- Fundacion MEDINA Parque tecnológico ciencias de la salud, Granada, Spain
| | | | | | | |
Collapse
|
50
|
Monecke T, Dickmanns A, Ficner R. Allosteric control of the exportin CRM1 unraveled by crystal structure analysis. FEBS J 2014; 281:4179-94. [PMID: 24823279 PMCID: PMC4231977 DOI: 10.1111/febs.12842] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2014] [Revised: 05/02/2014] [Accepted: 05/12/2014] [Indexed: 01/07/2023]
Abstract
Nucleocytoplasmic trafficking in eukaryotic cells is a highly regulated and coordinated process which involves an increasing variety of soluble nuclear transport receptors. Generally, transport receptors specifically bind their cargo and facilitate its transition through nuclear pore complexes, aqueous channels connecting the two compartments. Directionality of such transport events by receptors of the importin β superfamily requires the interaction with the small GTPase Ras-related nuclear antigen (Ran). While importins need RanGTP to release their cargo in the nucleus and thus to terminate import, exportins recruit cargo in the RanGTP-bound state. The exportin chromosome region maintenance 1 (CRM1) is a highly versatile transport receptor that exports a plethora of different protein and RNP cargoes. Moreover, binding of RanGTP and of cargo to CRM1 are highly cooperative events despite the fact that cargo and RanGTP do not interact directly in crystal structures of assembled export complexes. Integrative approaches have recently unraveled the individual steps of the CRM1 transport cycle at a structural level and explained how the HEAT-repeat architecture of CRM1 provides a framework for the key elements to mediate allosteric interactions with RanGTP, Ran binding proteins and cargo. Moreover, during the last decade, CRM1 has become a more and more appreciated target for anti-cancer drugs. Hence, detailed understanding of the flexibility, the regulatory features and the positive binding cooperativity between CRM1, Ran and cargo is a prerequisite for the development of highly effective drugs. Here we review recent structural advances in the characterization of CRM1 and CRM1-containing complexes with a special emphasis on X-ray crystallographic studies.
Collapse
Affiliation(s)
- Thomas Monecke
- Abteilung für Molekulare Strukturbiologie, Institut für Mikrobiologie und Genetik, Göttinger Zentrum für Molekulare Biowissenschaften, Georg-August-Universität Göttingen, Germany
| | | | | |
Collapse
|