1
|
Pellarin I, Dall'Acqua A, Favero A, Segatto I, Rossi V, Crestan N, Karimbayli J, Belletti B, Baldassarre G. Cyclin-dependent protein kinases and cell cycle regulation in biology and disease. Signal Transduct Target Ther 2025; 10:11. [PMID: 39800748 PMCID: PMC11734941 DOI: 10.1038/s41392-024-02080-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 10/16/2024] [Accepted: 11/13/2024] [Indexed: 01/18/2025] Open
Abstract
Cyclin Dependent Kinases (CDKs) are closely connected to the regulation of cell cycle progression, having been first identified as the kinases able to drive cell division. In reality, the human genome contains 20 different CDKs, which can be divided in at least three different sub-family with different functions, mechanisms of regulation, expression patterns and subcellular localization. Most of these kinases play fundamental roles the normal physiology of eucaryotic cells; therefore, their deregulation is associated with the onset and/or progression of multiple human disease including but not limited to neoplastic and neurodegenerative conditions. Here, we describe the functions of CDKs, categorized into the three main functional groups in which they are classified, highlighting the most relevant pathways that drive their expression and functions. We then discuss the potential roles and deregulation of CDKs in human pathologies, with a particular focus on cancer, the human disease in which CDKs have been most extensively studied and explored as therapeutic targets. Finally, we discuss how CDKs inhibitors have become standard therapies in selected human cancers and propose novel ways of investigation to export their targeting from cancer to other relevant chronic diseases. We hope that the effort we made in collecting all available information on both the prominent and lesser-known CDK family members will help in identify and develop novel areas of research to improve the lives of patients affected by debilitating chronic diseases.
Collapse
Affiliation(s)
- Ilenia Pellarin
- Division of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, National Cancer Institute, Aviano, Italy
| | - Alessandra Dall'Acqua
- Division of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, National Cancer Institute, Aviano, Italy
| | - Andrea Favero
- Division of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, National Cancer Institute, Aviano, Italy
| | - Ilenia Segatto
- Division of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, National Cancer Institute, Aviano, Italy
| | - Valentina Rossi
- Division of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, National Cancer Institute, Aviano, Italy
| | - Nicole Crestan
- Division of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, National Cancer Institute, Aviano, Italy
| | - Javad Karimbayli
- Division of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, National Cancer Institute, Aviano, Italy
| | - Barbara Belletti
- Division of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, National Cancer Institute, Aviano, Italy
| | - Gustavo Baldassarre
- Division of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, National Cancer Institute, Aviano, Italy.
| |
Collapse
|
2
|
Gupta H, Lee JR, Hoffman KB, Min KW, Yoon JH. RNA Decay Assay: 5-Ethynyl-Uridine Labeling and Chasing. Methods Mol Biol 2025; 2863:139-149. [PMID: 39535709 DOI: 10.1007/978-1-0716-4176-7_10] [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] [Indexed: 11/16/2024]
Abstract
Eukaryotic RNA synthesis and degradation are intricately regulated, impacting on gene expression dynamics. RNA stability varies in individual transcripts and is modulated by trans-acting factors such as microRNAs, long noncoding RNAs, and RNA-binding proteins, which determine protein output and subsequent cellular processes. To measure RNA decay rate, accurate and reliable methodologies are essential in the field of RNA biology. Transcription inhibition and metabolic labeling enable comprehensive investigations on RNA decay, offering critical insights into dynamic regulation of RNA decay. Transcription shut-off has been employed widely by using various approaches, such as treatment with chemical inhibitors or generation of temperature-sensitive mutants of RNA polymerases. However, it has limitations, providing a static view and lacking real-time dynamics as well as precise measurement of decay rate. Metabolic labeling, a method of incorporating modified nucleotides into RNA transcripts, complements shut-off approaches, allowing selective monitoring of newly synthesized RNA and tracing decay intermediates. The purpose of the protocol described in this chapter is to assess the kinetics and statics of newly synthesized RNA and its decay by 5-ethynyl uridine labeling.
Collapse
Affiliation(s)
- Haripriya Gupta
- Department of Oncology Science, University of Oklahoma, Oklahoma City, OK, USA
| | - Jayden R Lee
- Department of Oncology Science, University of Oklahoma, Oklahoma City, OK, USA
| | - Kane B Hoffman
- Department of Oncology Science, University of Oklahoma, Oklahoma City, OK, USA
| | - Kyung-Won Min
- Department of Biology, Gangneung-Wonju National University, Gangneung, Republic of Korea.
| | - Je-Hyun Yoon
- Department of Oncology Science, University of Oklahoma, Oklahoma City, OK, USA.
- Department of Pathology, University of Oklahoma, Oklahoma City, OK, USA.
| |
Collapse
|
3
|
Bedi K, Magnuson B, Narayanan IV, McShane A, Ashaka M, Paulsen MT, Wilson TE, Ljungman M. Isoform and pathway-specific regulation of post-transcriptional RNA processing in human cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.12.598705. [PMID: 38915566 PMCID: PMC11195214 DOI: 10.1101/2024.06.12.598705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
Steady-state levels of RNA transcripts are controlled by their rates of synthesis and degradation. Here we used nascent RNA Bru-seq and BruChase-seq to profile RNA dynamics across 16 human cell lines as part of ENCODE4 Deeply Profiled Cell Lines collection. We show that RNA turnover dynamics differ widely between transcripts of different genes and between different classes of RNA. Gene set enrichment analysis (GSEA) revealed that transcripts encoding proteins belonging to the same pathway often show similar turnover dynamics. Furthermore, transcript isoforms show distinct dynamics suggesting that RNA turnover is important in regulating mRNA isoform choice. Finally, splicing across newly made transcripts appears to be cooperative with either all or none type splicing. These data sets generated as part of ENCODE4 illustrate the intricate and coordinated regulation of RNA dynamics in controlling gene expression to allow for the precise coordination of cellular functions.
Collapse
Affiliation(s)
- Karan Bedi
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Biostatistics, School of Public Health, University of Michigan, Ann Arbor, MI 48109, USA
- Rogel Cancer Center and Center for RNA Biomedicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Brian Magnuson
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Pathology and Department of Human Genetics, University of Michigan Medical School, University of Michigan, Ann Arbor, MI 48109, USA
| | | | - Ariel McShane
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI 48109, USA
- Program in Cellular and Molecular Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Mario Ashaka
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Michelle T Paulsen
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Thomas E Wilson
- Rogel Cancer Center and Center for RNA Biomedicine, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Pathology and Department of Human Genetics, University of Michigan Medical School, University of Michigan, Ann Arbor, MI 48109, USA
| | - Mats Ljungman
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI 48109, USA
- Rogel Cancer Center and Center for RNA Biomedicine, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Environmental Health Sciences, School of Public Health, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
4
|
Yalala S, Gondane A, Poulose N, Liang J, Mills IG, Itkonen HM. CDK9 inhibition activates innate immune response through viral mimicry. FASEB J 2024; 38:e23628. [PMID: 38661032 DOI: 10.1096/fj.202302375r] [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: 11/19/2023] [Revised: 04/02/2024] [Accepted: 04/11/2024] [Indexed: 04/26/2024]
Abstract
Cancer cells frequently exhibit hyperactivation of transcription, which can lead to increased sensitivity to compounds targeting the transcriptional kinases, in particular CDK9. However, mechanistic details of CDK9 inhibition-induced cancer cell-selective anti-proliferative effects remain largely unknown. Here, we discover that CDK9 inhibition activates the innate immune response through viral mimicry in cancer cells. In MYC over-expressing prostate cancer cells, CDK9 inhibition leads to the gross accumulation of mis-spliced RNA. Double-stranded RNA (dsRNA)-activated kinase can recognize these mis-spliced RNAs, and we show that the activity of this kinase is required for the CDK9 inhibitor-induced anti-proliferative effects. Using time-resolved transcriptional profiling (SLAM-seq), targeted proteomics, and ChIP-seq, we show that, similar to viral infection, CDK9 inhibition significantly suppresses transcription of most genes but allows selective transcription and translation of cytokines related to the innate immune response. In particular, CDK9 inhibition activates NFκB-driven cytokine signaling at the transcriptional and secretome levels. The transcriptional signature induced by CDK9 inhibition identifies prostate cancers with a high level of genome instability. We propose that it is possible to induce similar effects in patients using CDK9 inhibition, which, we show, causes DNA damage in vitro. In the future, it is important to establish whether CDK9 inhibitors can potentiate the effects of immunotherapy against late-stage prostate cancer, a currently lethal disease.
Collapse
Affiliation(s)
- Shivani Yalala
- Department of Biochemistry and Developmental Biology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Aishwarya Gondane
- Department of Biochemistry and Developmental Biology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Ninu Poulose
- Nuffield Department of Surgical Sciences, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - Jing Liang
- Department of Biochemistry and Developmental Biology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Ian G Mills
- Nuffield Department of Surgical Sciences, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - Harri M Itkonen
- Department of Biochemistry and Developmental Biology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| |
Collapse
|
5
|
Burkart SS, Schweinoch D, Frankish J, Sparn C, Wüst S, Urban C, Merlo M, Magalhães VG, Piras A, Pichlmair A, Willemsen J, Kaderali L, Binder M. High-resolution kinetic characterization of the RIG-I-signaling pathway and the antiviral response. Life Sci Alliance 2023; 6:e202302059. [PMID: 37558422 PMCID: PMC10412806 DOI: 10.26508/lsa.202302059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 07/24/2023] [Accepted: 07/27/2023] [Indexed: 08/11/2023] Open
Abstract
RIG-I recognizes viral dsRNA and activates a cell-autonomous antiviral response. Upon stimulation, it triggers a signaling cascade leading to the production of type I and III IFNs. IFNs are secreted and signal to elicit the expression of IFN-stimulated genes, establishing an antiviral state of the cell. The topology of this pathway has been studied intensively, however, its exact dynamics are less understood. Here, we employed electroporation to synchronously activate RIG-I, enabling us to characterize cell-intrinsic innate immune signaling at a high temporal resolution. Employing IFNAR1/IFNLR-deficient cells, we could differentiate primary RIG-I signaling from secondary signaling downstream of the IFN receptors. Based on these data, we developed a comprehensive mathematical model capable of simulating signaling downstream of dsRNA recognition by RIG-I and the feedback and signal amplification by IFN. We further investigated the impact of viral antagonists on signaling dynamics. Our work provides a comprehensive insight into the signaling events that occur early upon virus infection and opens new avenues to study and disentangle the complexity of the host-virus interface.
Collapse
Affiliation(s)
- Sandy S Burkart
- Research Group "Dynamics of Early Viral Infection and the Innate Antiviral Response", Division Virus-Associated Carcinogenesis (F170), German Cancer Research Center, Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Darius Schweinoch
- Institute of Bioinformatics & Center for Functional Genomics of Microbes, University Medicine Greifswald, Greifswald, Germany
| | - Jamie Frankish
- Research Group "Dynamics of Early Viral Infection and the Innate Antiviral Response", Division Virus-Associated Carcinogenesis (F170), German Cancer Research Center, Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Carola Sparn
- Research Group "Dynamics of Early Viral Infection and the Innate Antiviral Response", Division Virus-Associated Carcinogenesis (F170), German Cancer Research Center, Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Sandra Wüst
- Research Group "Dynamics of Early Viral Infection and the Innate Antiviral Response", Division Virus-Associated Carcinogenesis (F170), German Cancer Research Center, Heidelberg, Germany
| | - Christian Urban
- Technical University of Munich, School of Medicine, Institute of Virology, Munich, Germany
| | - Marta Merlo
- Research Group "Dynamics of Early Viral Infection and the Innate Antiviral Response", Division Virus-Associated Carcinogenesis (F170), German Cancer Research Center, Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Vladimir G Magalhães
- Research Group "Dynamics of Early Viral Infection and the Innate Antiviral Response", Division Virus-Associated Carcinogenesis (F170), German Cancer Research Center, Heidelberg, Germany
| | - Antonio Piras
- Technical University of Munich, School of Medicine, Institute of Virology, Munich, Germany
| | - Andreas Pichlmair
- Technical University of Munich, School of Medicine, Institute of Virology, Munich, Germany
- German Center for Infection Research (DZIF), Munich Partner Site, Munich, Germany
| | - Joschka Willemsen
- Research Group "Dynamics of Early Viral Infection and the Innate Antiviral Response", Division Virus-Associated Carcinogenesis (F170), German Cancer Research Center, Heidelberg, Germany
| | - Lars Kaderali
- Institute of Bioinformatics & Center for Functional Genomics of Microbes, University Medicine Greifswald, Greifswald, Germany
| | - Marco Binder
- Research Group "Dynamics of Early Viral Infection and the Innate Antiviral Response", Division Virus-Associated Carcinogenesis (F170), German Cancer Research Center, Heidelberg, Germany
| |
Collapse
|
6
|
Jiang L, Wen C, Zhou H, Liu A, Zhang H, Chen X, Ding W, Liu J, Shi X. Cyclin-dependent kinase 7/9 inhibitor SNS-032 induces apoptosis in diffuse large B-cell lymphoma cells. Cancer Biol Ther 2022; 23:319-327. [PMID: 35332847 PMCID: PMC8959513 DOI: 10.1080/15384047.2022.2055421] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Approximately 40% of patients with diffuse large B-cell lymphoma (DLBCL) are refractory or relapse to standard chemotherapy, and most of them are activated B cell-like DLBCLs (ABC-DLBCL) and germinal center B cell-like DLBCLs (GCB-DLBCL). SNS-032, a novel and selective CDK7/9 inhibitor, that the first phase clinical trials approved by US FDA for cancer treatment have been completed. In this study, we investigated the anti-tumor effect of SNS-032 in ABC- and GCB-DLBCL subtypes. We report that SNS-032 induced growth inhibition and cell apoptosis in both DLBCL cells in vitro, and inhibited the growth of both DLBCL xenografts in nude mice. Mechanistically, SNS-032 inhibited RNA polymerase II, which led to transcriptional-dependent suppression of NF-κB signaling pathway and its downstream targets involved in cell survival; SNS-032 also downregulates BCL-2 and c-MYC in both mRNA and protein levels. Significantly, these findings provide pre-clinical evidence for application of targeting the CDK7/9 in DLBCL.
Collapse
Affiliation(s)
- Liling Jiang
- The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital; Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradatio, State Key Laboratory of Respiratory Disease; Affiliated Cancer Hospital of Guangzhou Medical University; School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou P.R. China
| | - Chuangyu Wen
- Department of Obstetrics and Gynaecology, Dongguan Affiliated Hospital, Southern Medical University, Dongguan, P.R. China
| | - Huan Zhou
- The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital; Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradatio, State Key Laboratory of Respiratory Disease; Affiliated Cancer Hospital of Guangzhou Medical University; School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou P.R. China
| | - Aochu Liu
- The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital; Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradatio, State Key Laboratory of Respiratory Disease; Affiliated Cancer Hospital of Guangzhou Medical University; School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou P.R. China
| | - Haichuan Zhang
- The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital; Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradatio, State Key Laboratory of Respiratory Disease; Affiliated Cancer Hospital of Guangzhou Medical University; School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou P.R. China
| | - Xinmei Chen
- The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital; Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradatio, State Key Laboratory of Respiratory Disease; Affiliated Cancer Hospital of Guangzhou Medical University; School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou P.R. China
| | - Wa Ding
- The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital; Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradatio, State Key Laboratory of Respiratory Disease; Affiliated Cancer Hospital of Guangzhou Medical University; School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou P.R. China
| | - Jinbao Liu
- The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital; Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradatio, State Key Laboratory of Respiratory Disease; Affiliated Cancer Hospital of Guangzhou Medical University; School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou P.R. China
| | - Xianping Shi
- The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital; Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradatio, State Key Laboratory of Respiratory Disease; Affiliated Cancer Hospital of Guangzhou Medical University; School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou P.R. China
| |
Collapse
|
7
|
Agarwal V, Kelley DR. The genetic and biochemical determinants of mRNA degradation rates in mammals. Genome Biol 2022; 23:245. [PMID: 36419176 PMCID: PMC9684954 DOI: 10.1186/s13059-022-02811-x] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 11/02/2022] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND Degradation rate is a fundamental aspect of mRNA metabolism, and the factors governing it remain poorly characterized. Understanding the genetic and biochemical determinants of mRNA half-life would enable more precise identification of variants that perturb gene expression through post-transcriptional gene regulatory mechanisms. RESULTS We establish a compendium of 39 human and 27 mouse transcriptome-wide mRNA decay rate datasets. A meta-analysis of these data identified a prevalence of technical noise and measurement bias, induced partially by the underlying experimental strategy. Correcting for these biases allowed us to derive more precise, consensus measurements of half-life which exhibit enhanced consistency between species. We trained substantially improved statistical models based upon genetic and biochemical features to better predict half-life and characterize the factors molding it. Our state-of-the-art model, Saluki, is a hybrid convolutional and recurrent deep neural network which relies only upon an mRNA sequence annotated with coding frame and splice sites to predict half-life (r=0.77). The key novel principle learned by Saluki is that the spatial positioning of splice sites, codons, and RNA-binding motifs within an mRNA is strongly associated with mRNA half-life. Saluki predicts the impact of RNA sequences and genetic mutations therein on mRNA stability, in agreement with functional measurements derived from massively parallel reporter assays. CONCLUSIONS Our work produces a more robust ground truth for transcriptome-wide mRNA half-lives in mammalian cells. Using these revised measurements, we trained Saluki, a model that is over 50% more accurate in predicting half-life from sequence than existing models. Saluki succinctly captures many of the known determinants of mRNA half-life and can be rapidly deployed to predict the functional consequences of arbitrary mutations in the transcriptome.
Collapse
Affiliation(s)
- Vikram Agarwal
- Calico Life Sciences LLC, South San Francisco, CA, 94080, USA.
- Present Address: mRNA Center of Excellence, Sanofi Pasteur Inc., Waltham, MA, 02451, USA.
| | - David R Kelley
- Calico Life Sciences LLC, South San Francisco, CA, 94080, USA.
| |
Collapse
|
8
|
Rienecker KDA, Poston RG, Segales JS, Finholm IW, Sono MH, Munteanu SJ, Ghaninejad-Esfahani M, Rejepova A, Tejeda-Garibay S, Wickman K, Marron Fernandez de Velasco E, Thayer SA, Saha RN. Mild membrane depolarization in neurons induces immediate early gene transcription and acutely subdues responses to successive stimulus. J Biol Chem 2022; 298:102278. [PMID: 35863435 PMCID: PMC9396413 DOI: 10.1016/j.jbc.2022.102278] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Revised: 07/12/2022] [Accepted: 07/13/2022] [Indexed: 11/22/2022] Open
Abstract
Immediate early genes (IEGs) are transcribed in response to neuronal activity from sensory stimulation during multiple adaptive processes in the brain. The transcriptional profile of IEGs is indicative of the duration of neuronal activity, but its sensitivity to the strength of depolarization remains unknown. Also unknown is whether activity history of graded potential changes influence future neuronal activity. In this work with dissociated rat cortical neurons, we found that mild depolarization—mediated by elevated extracellular potassium (K+)—induces a wide array of rapid IEGs and transiently depresses transcriptional and signaling responses to a successive stimulus. This latter effect was independent of de novo transcription, translation, and signaling via calcineurin or mitogen-activated protein kinase. Furthermore, as measured by multiple electrode arrays and calcium imaging, mild depolarization acutely subdues subsequent spontaneous and bicuculline-evoked activity via calcium- and N-methyl-d-aspartate receptor–dependent mechanisms. Collectively, this work suggests that a recent history of graded potential changes acutely depress neuronal intrinsic properties and subsequent responses. Such effects may have several potential downstream implications, including reducing signal-to-noise ratio during synaptic plasticity processes.
Collapse
Affiliation(s)
- Kira D A Rienecker
- Molecular and Cell Biology Department, School of Natural Sciences, University of California, Merced, 5200 North Lake Road, Merced, California, USA, 95343
| | - Robert G Poston
- Molecular and Cell Biology Department, School of Natural Sciences, University of California, Merced, 5200 North Lake Road, Merced, California, USA, 95343
| | - Joshua S Segales
- Molecular and Cell Biology Department, School of Natural Sciences, University of California, Merced, 5200 North Lake Road, Merced, California, USA, 95343
| | - Isabelle W Finholm
- Department of Pharmacology, University of Minnesota Medical School, Minneapolis, MN, 55455, USA
| | - Morgan H Sono
- Molecular and Cell Biology Department, School of Natural Sciences, University of California, Merced, 5200 North Lake Road, Merced, California, USA, 95343
| | - Sorina J Munteanu
- Molecular and Cell Biology Department, School of Natural Sciences, University of California, Merced, 5200 North Lake Road, Merced, California, USA, 95343
| | - Mina Ghaninejad-Esfahani
- Molecular and Cell Biology Department, School of Natural Sciences, University of California, Merced, 5200 North Lake Road, Merced, California, USA, 95343
| | - Ayna Rejepova
- Molecular and Cell Biology Department, School of Natural Sciences, University of California, Merced, 5200 North Lake Road, Merced, California, USA, 95343
| | - Susana Tejeda-Garibay
- Molecular and Cell Biology Department, School of Natural Sciences, University of California, Merced, 5200 North Lake Road, Merced, California, USA, 95343
| | - Kevin Wickman
- Department of Pharmacology, University of Minnesota Medical School, Minneapolis, MN, 55455, USA
| | | | - Stanley A Thayer
- Department of Pharmacology, University of Minnesota Medical School, Minneapolis, MN, 55455, USA
| | - Ramendra N Saha
- Molecular and Cell Biology Department, School of Natural Sciences, University of California, Merced, 5200 North Lake Road, Merced, California, USA, 95343.
| |
Collapse
|
9
|
Kim HJ, Booth G, Saunders L, Srivatsan S, McFaline-Figueroa JL, Trapnell C. Nuclear oligo hashing improves differential analysis of single-cell RNA-seq. Nat Commun 2022; 13:2666. [PMID: 35562344 PMCID: PMC9106741 DOI: 10.1038/s41467-022-30309-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Accepted: 04/26/2022] [Indexed: 11/09/2022] Open
Abstract
Single-cell RNA sequencing (scRNA-seq) offers a high-resolution molecular view into complex tissues, but suffers from high levels of technical noise which frustrates efforts to compare the gene expression programs of different cell types. "Spike-in" RNA standards help control for technical variation in scRNA-seq, but using them with recently developed, ultra-scalable scRNA-seq methods based on combinatorial indexing is not feasible. Here, we describe a simple and cost-effective method for normalizing transcript counts and subtracting technical variability that improves differential expression analysis in scRNA-seq. The method affixes a ladder of synthetic single-stranded DNA oligos to each cell that appears in its RNA-seq library. With improved normalization we explore chemical perturbations with broad or highly specific effects on gene regulation, including RNA pol II elongation, histone deacetylation, and activation of the glucocorticoid receptor. Our methods reveal that inhibiting histone deacetylation prevents cells from executing their canonical program of changes following glucocorticoid stimulation.
Collapse
Affiliation(s)
- Hyeon-Jin Kim
- Department of Genome Sciences, University of Washington, Seattle, WA, 98195, USA
| | - Greg Booth
- Department of Genome Sciences, University of Washington, Seattle, WA, 98195, USA
| | - Lauren Saunders
- Department of Genome Sciences, University of Washington, Seattle, WA, 98195, USA
| | - Sanjay Srivatsan
- Department of Genome Sciences, University of Washington, Seattle, WA, 98195, USA
| | | | - Cole Trapnell
- Department of Genome Sciences, University of Washington, Seattle, WA, 98195, USA. .,Brotman Baty Institute of Precision Medicine, Seattle, WA, 98195, USA. .,Allen Discovery Center for Cell Lineage Tracing, Seattle, WA, 98195, USA.
| |
Collapse
|
10
|
Khanjani F, Jafari L, Azadiyan S, Roozbehi S, Moradian C, Zahiri J, Hasannia S, Sajedi RH. Drug repositioning based on gene expression data for human HER2-positive breast cancer. Arch Biochem Biophys 2021; 712:109043. [PMID: 34597657 DOI: 10.1016/j.abb.2021.109043] [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: 05/26/2021] [Revised: 09/09/2021] [Accepted: 09/21/2021] [Indexed: 10/20/2022]
Abstract
Human epidermal growth factor receptor 2 (HER2)-positive breast cancer represents approximately 15-30% of all invasive breast cancers. Despite the recent advances in therapeutic practices of HER2 subtype, drug resistance and tumor recurrence still have remained as major problems. Drug discovery is a long and difficult process, so the aim of this study is to find potential new application for existing therapeutic agents. Gene expression data for breast invasive carcinoma were retrieved from The Cancer Genome Atlas (TCGA) database. The normal and tumor samples were analyzed using Linear Models for Microarray Data (LIMMA) R package in order to find the differentially expressed genes (DEGs). These genes were used as entry for the library of integrated network-based cellular signatures (LINCS) L1000CDS2 software and suggested 24 repurposed drugs. According to the obtained results, some of these drugs including vorinostat, mocetinostat, alvocidib, CGP-60474, BMS-387032, AT-7519, and curcumin have significant functional similarity and structural correlation with FDA-approved breast cancer drugs. Based on the drug-target network, which consisted of the repurposed drugs and their target genes, the aforementioned drugs had the highest degrees. Moreover, the experimental approach verified curcumin as an effective therapeutic agent for HER2 positive breast cancer. Hence, our work suggested that some repurposed drugs based on gene expression data can be noticed as potential drugs for the treatment of HER2-positive breast cancer.
Collapse
Affiliation(s)
- Farkhondeh Khanjani
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Leila Jafari
- Department of Computer Science and Information Technology, Institute for Advanced Studies in Basic Sciences, Zanjan, Iran
| | - Somayeh Azadiyan
- Bioinformatics and Computational Omics Lab (BioCOOL), Department of Biophysics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Sahar Roozbehi
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Cobra Moradian
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Javad Zahiri
- Department of Pediatrics, University of California San Diego, La Jolla, CA, USA.
| | - Sadegh Hasannia
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Reza H Sajedi
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran.
| |
Collapse
|
11
|
Hu Q, Poulose N, Girmay S, Helevä A, Doultsinos D, Gondane A, Steele RE, Liu X, Loda M, Liu S, Tang DG, Mills IG, Itkonen HM. Inhibition of CDK9 activity compromises global splicing in prostate cancer cells. RNA Biol 2021; 18:722-729. [PMID: 34592899 PMCID: PMC8782174 DOI: 10.1080/15476286.2021.1983287] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Revised: 07/07/2021] [Accepted: 09/15/2021] [Indexed: 10/20/2022] Open
Abstract
Cyclin-dependent kinase 9 (CDK9) phosphorylates RNA polymerase II to promote productive transcription elongation. Here we show that short-term CDK9 inhibition affects the splicing of thousands of mRNAs. CDK9 inhibition impairs global splicing and there is no evidence for a coordinated response between the alternative splicing and the overall transcriptome. Alternative splicing is a feature of aggressive prostate cancer (CRPC) and enables the generation of the anti-androgen resistant version of the ligand-independent androgen receptor, AR-v7. We show that CDK9 inhibition results in the loss of AR and AR-v7 expression due to the defects in splicing, which sensitizes CRPC cells to androgen deprivation. Finally, we demonstrate that CDK9 expression increases as PC cells develop CRPC-phenotype both in vitro and also in patient samples. To conclude, here we show that CDK9 inhibition compromises splicing in PC cells, which can be capitalized on by targeting the PC-specific addiction androgen receptor.
Collapse
Affiliation(s)
- Qiang Hu
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Ninu Poulose
- Nuffield Department of Surgical Sciences, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - Samuel Girmay
- Department of Biochemistry and Developmental Biology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Alma Helevä
- Department of Biochemistry and Developmental Biology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Dimitrios Doultsinos
- Nuffield Department of Surgical Sciences, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - Aishwarya Gondane
- Department of Biochemistry and Developmental Biology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Rebecca E Steele
- PCUK/Movember Centre of Excellence for Prostate Cancer Research, Centre for Cancer Research and Cell Biology (CCRCB), Queen’s University Belfast, Belfast, UK
- Breast Cancer Now Toby Robins Research Centre, the Institute of Cancer Research, London, UK
| | - Xiaozhuo Liu
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Massimo Loda
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York-Presbyterian Hospital, New York, New York, USA
- The Broad Institute of Harvard and Mit, Cambridge, Massachusetts
- The New York Genome Center, New York, New York, USA
| | - Song Liu
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Dean G. Tang
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Ian G Mills
- Nuffield Department of Surgical Sciences, University of Oxford, John Radcliffe Hospital, Oxford, UK
- PCUK/Movember Centre of Excellence for Prostate Cancer Research, Centre for Cancer Research and Cell Biology (CCRCB), Queen’s University Belfast, Belfast, UK
| | - Harri M Itkonen
- Department of Biochemistry and Developmental Biology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| |
Collapse
|
12
|
Lau B, Kerr K, Camiolo S, Nightingale K, Gu Q, Antrobus R, Suárez NM, Loney C, Stanton RJ, Weekes MP, Davison AJ. Human Cytomegalovirus RNA2.7 Is Required for Upregulating Multiple Cellular Genes To Promote Cell Motility and Viral Spread Late in Lytic Infection. J Virol 2021; 95:e0069821. [PMID: 34346763 PMCID: PMC8475523 DOI: 10.1128/jvi.00698-21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Accepted: 07/22/2021] [Indexed: 11/20/2022] Open
Abstract
Long noncoding RNAs (lncRNAs) are frequently associated with broad modulation of gene expression and thus provide the cell with the ability to synchronize entire metabolic processes. We used transcriptomic approaches to investigate whether the most abundant human cytomegalovirus-encoded lncRNA, RNA2.7, has this characteristic. By comparing cells infected with wild-type virus (WT) to cells infected with RNA2.7 deletion mutants, RNA2.7 was implicated in regulating a large number of cellular genes late in lytic infection. Pathway analysis indicated that >100 of these genes are associated with promoting cell movement, and the 10 most highly regulated of these were validated in further experiments. Morphological analysis and live cell tracking of WT- and RNA2.7 mutant-infected cells indicated that RNA2.7 is involved in promoting the movement and detachment of infected cells late in infection, and plaque assays using sparse cell monolayers indicated that RNA2.7 is also involved in promoting cell-to-cell spread of virus. Consistent with the observation that upregulated mRNAs are relatively A+U-rich, which is a trait associated with transcript instability, and that they are also enriched in motifs associated with mRNA instability, transcriptional inhibition experiments on WT- and RNA2.7 mutant-infected cells showed that four upregulated transcripts lived longer in the presence of RNA2.7. These findings demonstrate that RNA2.7 is required for promoting cell movement and viral spread late in infection and suggest that this may be due to general stabilization of A+U-rich transcripts. IMPORTANCE In addition to messenger RNAs (mRNAs), the human genome encodes a large number of long noncoding RNAs (lncRNAs). Many lncRNAs that have been studied in detail are associated with broad modulation of gene expression and have important biological roles. Human cytomegalovirus, which is a large, clinically important DNA virus, specifies four lncRNAs, one of which (RNA2.7) is expressed at remarkably high levels during lytic infection. Our studies show that RNA2.7 is required for upregulating a large number of human genes, about 100 of which are associated with cell movement, and for promoting the movement of infected cells and the spread of virus from one cell to another. Further bioinformatic and experimental analyses indicated that RNA2.7 may exert these effects by stabilizing mRNAs that are relatively rich in A and U nucleotides. These findings increase our knowledge of how human cytomegalovirus regulates the infected cell to promote its own success.
Collapse
Affiliation(s)
- Betty Lau
- MRC-University of Glasgow Centre for Virus Research, Glasgow, United Kingdom
| | - Karen Kerr
- MRC-University of Glasgow Centre for Virus Research, Glasgow, United Kingdom
| | - Salvatore Camiolo
- MRC-University of Glasgow Centre for Virus Research, Glasgow, United Kingdom
| | - Katie Nightingale
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom
| | - Quan Gu
- MRC-University of Glasgow Centre for Virus Research, Glasgow, United Kingdom
| | - Robin Antrobus
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom
| | - Nicolás M. Suárez
- MRC-University of Glasgow Centre for Virus Research, Glasgow, United Kingdom
| | - Colin Loney
- MRC-University of Glasgow Centre for Virus Research, Glasgow, United Kingdom
| | - Richard J. Stanton
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - Michael P. Weekes
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom
| | - Andrew J. Davison
- MRC-University of Glasgow Centre for Virus Research, Glasgow, United Kingdom
| |
Collapse
|
13
|
Furlan M, de Pretis S, Pelizzola M. Dynamics of transcriptional and post-transcriptional regulation. Brief Bioinform 2021; 22:bbaa389. [PMID: 33348360 PMCID: PMC8294512 DOI: 10.1093/bib/bbaa389] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 11/12/2020] [Accepted: 11/27/2020] [Indexed: 02/07/2023] Open
Abstract
Despite gene expression programs being notoriously complex, RNA abundance is usually assumed as a proxy for transcriptional activity. Recently developed approaches, able to disentangle transcriptional and post-transcriptional regulatory processes, have revealed a more complex scenario. It is now possible to work out how synthesis, processing and degradation kinetic rates collectively determine the abundance of each gene's RNA. It has become clear that the same transcriptional output can correspond to different combinations of the kinetic rates. This underscores the fact that markedly different modes of gene expression regulation exist, each with profound effects on a gene's ability to modulate its own expression. This review describes the development of the experimental and computational approaches, including RNA metabolic labeling and mathematical modeling, that have been disclosing the mechanisms underlying complex transcriptional programs. Current limitations and future perspectives in the field are also discussed.
Collapse
Affiliation(s)
- Mattia Furlan
- Center for Genomic Science of IIT@SEMM, Fondazione Istituto Italiano di Tecnologia (IIT), 20139 Milan, Italy
| | - Stefano de Pretis
- Center for Genomic Science of IIT@SEMM, Fondazione Istituto Italiano di Tecnologia (IIT), 20139 Milan, Italy
| | - Mattia Pelizzola
- Center for Genomic Science of IIT@SEMM, Fondazione Istituto Italiano di Tecnologia (IIT), 20139 Milan, Italy
| |
Collapse
|
14
|
Shirai Y, Chow CCT, Kambe G, Suwa T, Kobayashi M, Takahashi I, Harada H, Nam JM. An Overview of the Recent Development of Anticancer Agents Targeting the HIF-1 Transcription Factor. Cancers (Basel) 2021; 13:cancers13112813. [PMID: 34200019 PMCID: PMC8200185 DOI: 10.3390/cancers13112813] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 05/30/2021] [Accepted: 05/31/2021] [Indexed: 02/07/2023] Open
Abstract
Hypoxia, a characteristic feature of solid tumors, is associated with the malignant phenotype and therapy resistance of cancers. Hypoxia-inducible factor 1 (HIF-1), which is responsible for the metazoan adaptive response to hypoxia, has been recognized as a rational target for cancer therapy due to its critical functions in hypoxic regions. In order to efficiently inhibit its activity, extensive efforts have been made to elucidate the molecular mechanism underlying the activation of HIF-1. Here, we provide an overview of relevant research, particularly on a series of HIF-1 activators identified so far and the development of anticancer drugs targeting them.
Collapse
Affiliation(s)
- Yukari Shirai
- Laboratory of Cancer Cell Biology, Graduate School of Biostudies, Kyoto University, Yoshida-Konoe-Cho, Sakyo-Ku, Kyoto 606-8501, Japan; (Y.S.); (C.C.T.C.); (G.K.); (T.S.); (M.K.); (I.T.)
- Department of Genome Repair Dynamics, Radiation Biology Center, Graduate School of Biostudies, Kyoto University, Yoshida-Konoe-Cho, Sakyo-Ku, Kyoto 606-8501, Japan
| | - Christalle C. T. Chow
- Laboratory of Cancer Cell Biology, Graduate School of Biostudies, Kyoto University, Yoshida-Konoe-Cho, Sakyo-Ku, Kyoto 606-8501, Japan; (Y.S.); (C.C.T.C.); (G.K.); (T.S.); (M.K.); (I.T.)
- Department of Genome Repair Dynamics, Radiation Biology Center, Graduate School of Biostudies, Kyoto University, Yoshida-Konoe-Cho, Sakyo-Ku, Kyoto 606-8501, Japan
| | - Gouki Kambe
- Laboratory of Cancer Cell Biology, Graduate School of Biostudies, Kyoto University, Yoshida-Konoe-Cho, Sakyo-Ku, Kyoto 606-8501, Japan; (Y.S.); (C.C.T.C.); (G.K.); (T.S.); (M.K.); (I.T.)
- Department of Genome Repair Dynamics, Radiation Biology Center, Graduate School of Biostudies, Kyoto University, Yoshida-Konoe-Cho, Sakyo-Ku, Kyoto 606-8501, Japan
| | - Tatsuya Suwa
- Laboratory of Cancer Cell Biology, Graduate School of Biostudies, Kyoto University, Yoshida-Konoe-Cho, Sakyo-Ku, Kyoto 606-8501, Japan; (Y.S.); (C.C.T.C.); (G.K.); (T.S.); (M.K.); (I.T.)
- Department of Genome Repair Dynamics, Radiation Biology Center, Graduate School of Biostudies, Kyoto University, Yoshida-Konoe-Cho, Sakyo-Ku, Kyoto 606-8501, Japan
| | - Minoru Kobayashi
- Laboratory of Cancer Cell Biology, Graduate School of Biostudies, Kyoto University, Yoshida-Konoe-Cho, Sakyo-Ku, Kyoto 606-8501, Japan; (Y.S.); (C.C.T.C.); (G.K.); (T.S.); (M.K.); (I.T.)
- Department of Genome Repair Dynamics, Radiation Biology Center, Graduate School of Biostudies, Kyoto University, Yoshida-Konoe-Cho, Sakyo-Ku, Kyoto 606-8501, Japan
| | - Itsuki Takahashi
- Laboratory of Cancer Cell Biology, Graduate School of Biostudies, Kyoto University, Yoshida-Konoe-Cho, Sakyo-Ku, Kyoto 606-8501, Japan; (Y.S.); (C.C.T.C.); (G.K.); (T.S.); (M.K.); (I.T.)
- Department of Genome Repair Dynamics, Radiation Biology Center, Graduate School of Biostudies, Kyoto University, Yoshida-Konoe-Cho, Sakyo-Ku, Kyoto 606-8501, Japan
| | - Hiroshi Harada
- Laboratory of Cancer Cell Biology, Graduate School of Biostudies, Kyoto University, Yoshida-Konoe-Cho, Sakyo-Ku, Kyoto 606-8501, Japan; (Y.S.); (C.C.T.C.); (G.K.); (T.S.); (M.K.); (I.T.)
- Department of Genome Repair Dynamics, Radiation Biology Center, Graduate School of Biostudies, Kyoto University, Yoshida-Konoe-Cho, Sakyo-Ku, Kyoto 606-8501, Japan
- Correspondence: (H.H.); (J.-M.N.); Tel.: +81-75-753-7560 (H.H.); +81-75-753-7567 (J.-M.N.)
| | - Jin-Min Nam
- Laboratory of Cancer Cell Biology, Graduate School of Biostudies, Kyoto University, Yoshida-Konoe-Cho, Sakyo-Ku, Kyoto 606-8501, Japan; (Y.S.); (C.C.T.C.); (G.K.); (T.S.); (M.K.); (I.T.)
- Department of Genome Repair Dynamics, Radiation Biology Center, Graduate School of Biostudies, Kyoto University, Yoshida-Konoe-Cho, Sakyo-Ku, Kyoto 606-8501, Japan
- Correspondence: (H.H.); (J.-M.N.); Tel.: +81-75-753-7560 (H.H.); +81-75-753-7567 (J.-M.N.)
| |
Collapse
|
15
|
Chen R, Tsai J, Thompson PA, Chen Y, Xiong P, Liu C, Burrows F, Sivina M, Burger JA, Keating MJ, Wierda WG, Plunkett W. The multi-kinase inhibitor TG02 induces apoptosis and blocks B-cell receptor signaling in chronic lymphocytic leukemia through dual mechanisms of action. Blood Cancer J 2021; 11:57. [PMID: 33714981 PMCID: PMC7956145 DOI: 10.1038/s41408-021-00436-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 01/13/2021] [Accepted: 01/25/2021] [Indexed: 02/06/2023] Open
Abstract
The constitutive activation of B-cell receptor (BCR) signaling, together with the overexpression of the Bcl-2 family anti-apoptotic proteins, represents two hallmarks of chronic lymphocytic leukemia (CLL) that drive leukemia cell proliferation and sustain their survival. TG02 is a small molecule multi-kinase inhibitor that simultaneously targets both of these facets of CLL pathogenesis. First, its inhibition of cyclin-dependent kinase 9 blocked the activation of RNA polymerase II and transcription. This led to the depletion of Mcl-1 and rapid induction of apoptosis in the primary CLL cells. This mechanism of apoptosis was independent of CLL prognostic factors or prior treatment history, but dependent on the expression of BAX and BAK. Second, TG02, which inhibits the members of the BCR signaling pathway such as Lck and Fyn, blocked BCR-crosslinking-induced activation of NF-κB and Akt, indicating abrogation of BCR signaling. Finally, the combination of TG02 and ibrutinib demonstrated moderate synergy, suggesting a future combination of TG02 with ibrutinib, or use in patients that are refractory to the BCR antagonists. Thus, the dual inhibitory activity on both the CLL survival pathway and BCR signaling identifies TG02 as a unique compound for clinical development in CLL and possibly other B cell malignancies.
Collapse
Affiliation(s)
- Rong Chen
- Department of Experimental Therapeutics, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA.
| | - Jennifer Tsai
- Department of Experimental Therapeutics, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA.,Department of Emergency Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Philip A Thompson
- Department of Leukemia, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Yuling Chen
- Department of Experimental Therapeutics, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Ping Xiong
- Department of Experimental Therapeutics, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Chaomei Liu
- Department of Experimental Therapeutics, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Francis Burrows
- Tragara Pharmaceuticals, Carlsbad, CA, USA.,Kura Oncology, Inc., San Diego, CA, USA
| | - Mariela Sivina
- Department of Leukemia, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Jan A Burger
- Department of Leukemia, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Michael J Keating
- Department of Leukemia, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - William G Wierda
- Department of Leukemia, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - William Plunkett
- Department of Experimental Therapeutics, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA.,Department of Leukemia, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
16
|
Blumberg A, Zhao Y, Huang YF, Dukler N, Rice EJ, Chivu AG, Krumholz K, Danko CG, Siepel A. Characterizing RNA stability genome-wide through combined analysis of PRO-seq and RNA-seq data. BMC Biol 2021; 19:30. [PMID: 33588838 PMCID: PMC7885420 DOI: 10.1186/s12915-021-00949-x] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 01/05/2021] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND The concentrations of distinct types of RNA in cells result from a dynamic equilibrium between RNA synthesis and decay. Despite the critical importance of RNA decay rates, current approaches for measuring them are generally labor-intensive, limited in sensitivity, and/or disruptive to normal cellular processes. Here, we introduce a simple method for estimating relative RNA half-lives that is based on two standard and widely available high-throughput assays: Precision Run-On sequencing (PRO-seq) and RNA sequencing (RNA-seq). RESULTS Our method treats PRO-seq as a measure of transcription rate and RNA-seq as a measure of RNA concentration, and estimates the rate of RNA decay required for a steady-state equilibrium. We show that this approach can be used to assay relative RNA half-lives genome-wide, with good accuracy and sensitivity for both coding and noncoding transcription units. Using a structural equation model (SEM), we test several features of transcription units, nearby DNA sequences, and nearby epigenomic marks for associations with RNA stability after controlling for their effects on transcription. We find that RNA splicing-related features are positively correlated with RNA stability, whereas features related to miRNA binding and DNA methylation are negatively correlated with RNA stability. Furthermore, we find that a measure based on U1 binding and polyadenylation sites distinguishes between unstable noncoding and stable coding transcripts but is not predictive of relative stability within the mRNA or lincRNA classes. We also identify several histone modifications that are associated with RNA stability. CONCLUSION We introduce an approach for estimating the relative half-lives of individual RNAs. Together, our estimation method and systematic analysis shed light on the pervasive impacts of RNA stability on cellular RNA concentrations.
Collapse
Affiliation(s)
- Amit Blumberg
- Simons Center for Quantitative Biology, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
| | - Yixin Zhao
- Simons Center for Quantitative Biology, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
| | - Yi-Fei Huang
- Simons Center for Quantitative Biology, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
- Present Address: Department of Biology and Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA, USA
| | - Noah Dukler
- Simons Center for Quantitative Biology, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
| | - Edward J Rice
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, Ithaca, NY, USA
| | - Alexandra G Chivu
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, Ithaca, NY, USA
| | - Katie Krumholz
- Simons Center for Quantitative Biology, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
| | - Charles G Danko
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, Ithaca, NY, USA
| | - Adam Siepel
- Simons Center for Quantitative Biology, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA.
| |
Collapse
|
17
|
Tian Y, Zeng Z, Li X, Wang Y, Chen R, Mattijssen S, Gaidamakov S, Wu Y, Maraia RJ, Peng W, Zhu J. Transcriptome-wide stability analysis uncovers LARP4-mediated NFκB1 mRNA stabilization during T cell activation. Nucleic Acids Res 2020; 48:8724-8739. [PMID: 32735645 PMCID: PMC7470963 DOI: 10.1093/nar/gkaa643] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 06/30/2020] [Accepted: 07/22/2020] [Indexed: 02/07/2023] Open
Abstract
T cell activation is a well-established model for studying cellular responses to exogenous stimulation. Motivated by our previous finding that intron retention (IR) could lead to transcript instability, in this study, we performed BruChase-Seq to experimentally monitor the expression dynamics of nascent transcripts in resting and activated CD4+ T cells. Computational modeling was then applied to quantify the stability of spliced and intron-retained transcripts on a genome-wide scale. Beyond substantiating that intron-retained transcripts were considerably less stable than spliced transcripts, we found a global stabilization of spliced mRNAs upon T cell activation, although the stability of intron-retained transcripts remained relatively constant. In addition, we identified that La-related protein 4 (LARP4), an RNA-binding protein (RBP) known to enhance mRNA stability, was involved in T cell activation-dependent mRNA stabilization. Knocking out Larp4 in mice destabilized Nfκb1 mRNAs and reduced secretion of interleukin-2 (IL2) and interferon-gamma (IFNγ), two factors critical for T cell proliferation and function. We propose that coordination between splicing regulation and mRNA stability may provide a novel paradigm to control spatiotemporal gene expression during T cell activation.
Collapse
Affiliation(s)
- Yi Tian
- Department of Physics, George Washington University, Washington, DC 20052, USA
- Systems Biology Center, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
- Institute of Immunology, PLA, Third Military Medical University, Chongqing 400038, PR China
| | - Zhouhao Zeng
- Department of Physics, George Washington University, Washington, DC 20052, USA
| | - Xiang Li
- Department of Physics, George Washington University, Washington, DC 20052, USA
| | - Yiyin Wang
- Systems Biology Center, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Runsen Chen
- Systems Biology Center, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
- Department of Cardiothoracic Surgery, Children's Hospital of Nanjing Medical University, Nanjing 210008, China
| | - Sandy Mattijssen
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Sergei Gaidamakov
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Yuzhang Wu
- Institute of Immunology, PLA, Third Military Medical University, Chongqing 400038, PR China
| | - Richard J Maraia
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Weiqun Peng
- Department of Physics, George Washington University, Washington, DC 20052, USA
| | - Jun Zhu
- Systems Biology Center, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
18
|
Therapeutic Targeting of the General RNA Polymerase II Transcription Machinery. Int J Mol Sci 2020; 21:ijms21093354. [PMID: 32397434 PMCID: PMC7246882 DOI: 10.3390/ijms21093354] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 05/06/2020] [Accepted: 05/07/2020] [Indexed: 12/27/2022] Open
Abstract
Inhibitors targeting the general RNA polymerase II (RNAPII) transcription machinery are candidate therapeutics in cancer and other complex diseases. Here, we review the molecular targets and mechanisms of action of these compounds, framing them within the steps of RNAPII transcription. We discuss the effects of transcription inhibitors in vitro and in cellular models (with an emphasis on cancer), as well as their efficacy in preclinical and clinical studies. We also discuss the rationale for inhibiting broadly acting transcriptional regulators or RNAPII itself in complex diseases.
Collapse
|
19
|
Cidado J, Boiko S, Proia T, Ferguson D, Criscione SW, San Martin M, Pop-Damkov P, Su N, Roamio Franklin VN, Sekhar Reddy Chilamakuri C, D'Santos CS, Shao W, Saeh JC, Koch R, Weinstock DM, Zinda M, Fawell SE, Drew L. AZD4573 Is a Highly Selective CDK9 Inhibitor That Suppresses MCL-1 and Induces Apoptosis in Hematologic Cancer Cells. Clin Cancer Res 2019; 26:922-934. [DOI: 10.1158/1078-0432.ccr-19-1853] [Citation(s) in RCA: 99] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 09/27/2019] [Accepted: 11/04/2019] [Indexed: 11/16/2022]
|
20
|
Lee DJ, Zeidner JF. Cyclin-dependent kinase (CDK) 9 and 4/6 inhibitors in acute myeloid leukemia (AML): a promising therapeutic approach. Expert Opin Investig Drugs 2019; 28:989-1001. [PMID: 31612739 DOI: 10.1080/13543784.2019.1678583] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Introduction: Despite advancements over the last 2 years, outcomes for acute myeloid leukemia (AML) are poor; however, a greater comprehension of disease mechanisms has driven the investigation of new targeted treatments. Cyclin-dependent kinases (CDKs) regulate cell cycle progression, transcription and DNA repair, and are aberrantly expressed in AML. Targeting the CDK pathway is an emerging promising therapeutic strategy in AML.Areas covered: We describe the rationale for targeting CDK9 and CDK4/6, the ongoing preclinical and clinical trials and the potential of these inhibitors in AML. Our analysis included an extensive literature search via the Pubmed database and clinicaltrials.gov (March to August, 2019).Expert opinion: While CDK4/6 inhibitors are early in development for AML, CDK9 inhibition with alvocidib has encouraging clinical activity in newly diagnosed and relapsed/refractory AML. Preclinical data suggests that leukemic MCL-1 dependence may predict response to alvocidib. Moreover, MCL-1 plays a key role in resistance to BCL-2 inhibition with venetoclax. Investigational strategies of concomitant BCL-2 and CDK9 inhibition represent a promising therapeutic platform for AML. Furthermore, preclinical data suggests that CDK4/6 inhibition has selective activity in patients with KMT2A-rearrangements and FLT3 mutations. Incorporation of CDK9 and 4/6 inhibitors into the existing therapeutic armamentarium may improve outcomes in AML.
Collapse
Affiliation(s)
- Daniel J Lee
- Department of Medicine, Division of Hematology/Oncology, Columbia University Irving Medical Center, New York, NY, USA
| | - Joshua F Zeidner
- Department of Medicine, Division of Hematology/Oncology, University of North Carolina, Lineberger Comprehensive Cancer Center, Chapel Hill, NC, USA
| |
Collapse
|
21
|
Schneeberger PE, Bierhals T, Neu A, Hempel M, Kutsche K. de novo MEPCE nonsense variant associated with a neurodevelopmental disorder causes disintegration of 7SK snRNP and enhanced RNA polymerase II activation. Sci Rep 2019; 9:12516. [PMID: 31467394 PMCID: PMC6715695 DOI: 10.1038/s41598-019-49032-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Accepted: 08/19/2019] [Indexed: 02/06/2023] Open
Abstract
In eukaryotes, the elongation phase of transcription by RNA polymerase II (RNAP II) is regulated by the transcription elongation factor b (P-TEFb), composed of Cyclin-T1 and cyclin-dependent kinase 9. The release of RNAP II is mediated by phosphorylation through P-TEFb that in turn is under control by the inhibitory 7SK small nuclear ribonucleoprotein (snRNP) complex. The 7SK snRNP consists of the 7SK non-coding RNA and the proteins MEPCE, LARP7, and HEXIM1/2. Biallelic LARP7 loss-of-function variants underlie Alazami syndrome characterized by growth retardation and intellectual disability. We report a boy with global developmental delay and seizures carrying the de novo MEPCE nonsense variant c.1552 C > T/p.(Arg518*). mRNA and protein analyses identified nonsense-mediated mRNA decay to underlie the decreased amount of MEPCE in patient fibroblasts followed by LARP7 and 7SK snRNA downregulation and HEXIM1 upregulation. Reduced binding of HEXIM1 to Cyclin-T1, hyperphosphorylation of the RNAP II C-terminal domain, and upregulated expression of ID2, ID3, MRPL11 and snRNAs U1, U2 and U4 in patient cells are suggestive of enhanced activation of P-TEFb. Flavopiridol treatment and ectopic MEPCE protein expression in patient fibroblasts rescued increased expression of six RNAP II-sensitive genes and suggested a possible repressive effect of MEPCE on P-TEFb-dependent transcription of specific genes.
Collapse
Affiliation(s)
- Pauline E Schneeberger
- Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Tatjana Bierhals
- Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Axel Neu
- Childrens Hospital, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Maja Hempel
- Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Kerstin Kutsche
- Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| |
Collapse
|
22
|
Rahaman MH, Lam F, Zhong L, Teo T, Adams J, Yu M, Milne RW, Pepper C, Lokman NA, Ricciardelli C, Oehler MK, Wang S. Targeting CDK9 for treatment of colorectal cancer. Mol Oncol 2019; 13:2178-2193. [PMID: 31398271 PMCID: PMC6763784 DOI: 10.1002/1878-0261.12559] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Revised: 07/30/2019] [Accepted: 08/07/2019] [Indexed: 12/11/2022] Open
Abstract
Colorectal cancer (CRC) remains one of the most lethal human malignancies, and pursuit of new therapeutic targets for treatment has been a major research focus. Cyclin-dependent kinase 9 (CDK9), which plays a crucial role in transcription, has emerged as a target for cancer treatment. CDKI-73, one of the most potent and pharmacologically superior CDK9 inhibitors, has demonstrated excellent anti-tumour efficacy against several types of cancers. In this study, we evaluated its therapeutic potential against CRC. CDKI-73 elicited high cytotoxicity against all colon cancer cell lines tested. Cell cycle and apoptosis analysis in HCT 116 and HT29 cells revealed that CDKI-73 induced cell death without accumulation of DNA at any phase of the cell cycle. Moreover, it caused depolarisation of mitochondrial membrane, leading to caspase-independent apoptosis. Knockdown by shRNA demonstrated the CDK9-targeted mechanism of CDKI-73, which also affected the Mnk/eIF4E signalling axis. In addition, RT-qPCR analysis showed that CDKI-73 down-regulated multiple pro-survival factors at the mRNA level. Its in vivo anti-tumour efficacy was further evaluated in Balb/c nude mice bearing HCT 116 xenograft tumours. CDKI-73 significantly inhibited tumour growth (***P < 0.001) without overt toxicity. Analysis of the tumour tissues collected from the xenografted animals confirmed that the in vivo anti-tumour efficacy was associated with CDK9 targeting of CDKI-73. Overall, this study provides compelling evidence that CDKI-73 is a promising drug candidate for treating colorectal cancer.
Collapse
Affiliation(s)
- Muhammed H Rahaman
- Centre for Drug Discovery and Development, School of Pharmacy and Medical Sciences, University of South Australia Cancer Research Institute, Adelaide, SA, Australia
| | - Frankie Lam
- Centre for Drug Discovery and Development, School of Pharmacy and Medical Sciences, University of South Australia Cancer Research Institute, Adelaide, SA, Australia
| | - Longjin Zhong
- Centre for Drug Discovery and Development, School of Pharmacy and Medical Sciences, University of South Australia Cancer Research Institute, Adelaide, SA, Australia
| | - Theodosia Teo
- Centre for Drug Discovery and Development, School of Pharmacy and Medical Sciences, University of South Australia Cancer Research Institute, Adelaide, SA, Australia
| | - Julian Adams
- Centre for Drug Discovery and Development, School of Pharmacy and Medical Sciences, University of South Australia Cancer Research Institute, Adelaide, SA, Australia
| | - Mingfeng Yu
- Centre for Drug Discovery and Development, School of Pharmacy and Medical Sciences, University of South Australia Cancer Research Institute, Adelaide, SA, Australia
| | - Robert W Milne
- Centre for Drug Discovery and Development, School of Pharmacy and Medical Sciences, University of South Australia Cancer Research Institute, Adelaide, SA, Australia
| | - Chris Pepper
- School of Medicine, Cardiff University, Health Park, UK
| | - Noor A Lokman
- Discipline of Obstetrics and Gynaecology, Adelaide Medical School, University of Adelaide, SA, Australia
| | - Carmela Ricciardelli
- Discipline of Obstetrics and Gynaecology, Adelaide Medical School, University of Adelaide, SA, Australia
| | - Martin K Oehler
- Discipline of Obstetrics and Gynaecology, Adelaide Medical School, University of Adelaide, SA, Australia
| | - Shudong Wang
- Centre for Drug Discovery and Development, School of Pharmacy and Medical Sciences, University of South Australia Cancer Research Institute, Adelaide, SA, Australia
| |
Collapse
|
23
|
Feng Y, Liu M, Wang Z, Zhao X, Han B, Xing Y, Wang M, Yang Y. A 4-bp deletion in the 5'UTR of TaAFP-B is associated with seed dormancy in common wheat (Triticum aestivum L.). BMC PLANT BIOLOGY 2019; 19:349. [PMID: 31399044 PMCID: PMC6688260 DOI: 10.1186/s12870-019-1950-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Accepted: 07/29/2019] [Indexed: 05/06/2023]
Abstract
BACKGROUND AFP is a negative regulator of ABA signaling that promotes ABI5 protein degradation and weakens regulation of ABA signaling by targeting upstream genes of ABI5, and TaABI5 gene was seed-specific, and accumulated during wheat grain maturation and dormancy acquisition, which played an important role in seed dormancy; TaAFP has a conserved domain with AFP, so TaAFP may also play an important role in seed dormancy in wheat. RESULTS Two allelic variants of TaAFP were identified on chromosome 2BS in common wheat, and designated as TaAFP-B1a and TaAFP-B1b. Sequence analysis showed a 4-bp deletion in the 5'UTR region of TaAFP-B1b compared with TaAFP-B1a. Based on the 4-bp deletion, a co-dominant functional marker of TaAFP-B was developed and designated as AFPB. The genotype generating a 203-bp fragment (TaAFP-B1b) was more resistant to pre-harvest sprouting than the genotype producing a 207-bp fragment (TaAFP-B1a) in a test of 91 white-grained Chinese wheat cultivars and advanced lines. The average germination index(GI) values of TaAFP-B1a and that of TaAFP-B1b were 45.18 and 30.72%, respectively, indicating a significant difference (P < 0.001). Moreover, the 4-bp deletion located in the 5'UTR not only affected the transcription level of TaAFP-B but also affected the mRNA decay, reduced the translation level of GUS and tdTomatoER and GUS activity in wheat leaves of transient expression. The transcript expression and the mRNA half-life value of TaAFP-B1a in developing seeds and mature seeds were much higher than those of TaAFP-B1b. CONCLUSION We identified a 4-bp InDel in the 5'UTR of TaAFP-B, which affected the mRNA transcription level, mRNA decay, translation levels of GUS and tdTomatoER, GUS activity, and was significantly associated with seed dormancy in common wheat. A functional marker was developed and validated based on this InDel.
Collapse
Affiliation(s)
- Yumei Feng
- College of Life Sciences, Inner Mongolia Agricultural University, Inner Mongolia Key Laboratory of Plant Stress Physiology and Molecular Biology, Erdos Road, Hohhot, 010018 Inner Mongolia China
| | - Meng Liu
- College of Life Sciences, Inner Mongolia Agricultural University, Inner Mongolia Key Laboratory of Plant Stress Physiology and Molecular Biology, Erdos Road, Hohhot, 010018 Inner Mongolia China
| | - Zeng Wang
- College of Life Sciences, Inner Mongolia Agricultural University, Inner Mongolia Key Laboratory of Plant Stress Physiology and Molecular Biology, Erdos Road, Hohhot, 010018 Inner Mongolia China
| | - Xianlin Zhao
- Wheat Research Institute, Henan Academy of Agricultural Sciences, Henan Key Laboratory of Wheat Biology, National Engineering Laboratory for Wheat, Key Laboratory of Wheat Biology and Genetic Breeding in Central Huang-Huai Region, Ministry of Agriculture, Zhengzhou, 450002 China
| | - Bing Han
- College of Life Sciences, Inner Mongolia Agricultural University, Inner Mongolia Key Laboratory of Plant Stress Physiology and Molecular Biology, Erdos Road, Hohhot, 010018 Inner Mongolia China
| | - Yanping Xing
- College of Life Sciences, Inner Mongolia Agricultural University, Inner Mongolia Key Laboratory of Plant Stress Physiology and Molecular Biology, Erdos Road, Hohhot, 010018 Inner Mongolia China
| | - Maoyan Wang
- College of Life Sciences, Inner Mongolia Agricultural University, Inner Mongolia Key Laboratory of Plant Stress Physiology and Molecular Biology, Erdos Road, Hohhot, 010018 Inner Mongolia China
| | - Yan Yang
- College of Life Sciences, Inner Mongolia Agricultural University, Inner Mongolia Key Laboratory of Plant Stress Physiology and Molecular Biology, Erdos Road, Hohhot, 010018 Inner Mongolia China
| |
Collapse
|
24
|
Senichkin VV, Streletskaia AY, Zhivotovsky B, Kopeina GS. Molecular Comprehension of Mcl-1: From Gene Structure to Cancer Therapy. Trends Cell Biol 2019; 29:549-562. [DOI: 10.1016/j.tcb.2019.03.004] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 03/14/2019] [Accepted: 03/19/2019] [Indexed: 01/19/2023]
|
25
|
Kelley CM, Ginsberg SD, Alldred MJ, Strupp BJ, Mufson EJ. Maternal Choline Supplementation Alters Basal Forebrain Cholinergic Neuron Gene Expression in the Ts65Dn Mouse Model of Down Syndrome. Dev Neurobiol 2019; 79:664-683. [PMID: 31120189 PMCID: PMC6756931 DOI: 10.1002/dneu.22700] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 05/16/2019] [Accepted: 05/18/2019] [Indexed: 12/12/2022]
Abstract
Down syndrome (DS), trisomy 21, is marked by intellectual disability and a premature aging profile including degeneration of the basal forebrain cholinergic neuron (BFCN) projection system, similar to Alzheimer's disease (AD). Although data indicate that perinatal maternal choline supplementation (MCS) alters the structure and function of these neurons in the Ts65Dn mouse model of DS and AD (Ts), whether MCS affects the molecular profile of vulnerable BFCNs remains unknown. We investigated the genetic signature of BFCNs obtained from Ts and disomic (2N) offspring of Ts65Dn dams maintained on a MCS diet (Ts+, 2N+) or a choline normal diet (ND) from mating until weaning, then maintained on ND until 4.4-7.5 months of age. Brains were then collected and prepared for choline acetyltransferase (ChAT) immunohistochemistry and laser capture microdissection followed by RNA extraction and custom-designed microarray analysis. Findings revealed upregulation of select transcripts in classes of genes related to the cytoskeleton (Tubb4b), AD (Cav1), cell death (Bcl2), presynaptic (Syngr1), immediate early (Fosb, Arc), G protein signaling (Gabarap, Rgs10), and cholinergic neurotransmission (Chrnb3) in Ts compared to 2N mice, which were normalized with MCS. Moreover, significant downregulation was seen in select transcripts associated with the cytoskeleton (Dync1h1), intracellular signaling (Itpka, Gng3, and Mlst8), and cell death (Ccng1) in Ts compared to 2N mice that was normalized with MCS. This study provides insight into genotype-dependent differences and the effects of MCS at the molecular level within a key vulnerable cell type in DS and AD.
Collapse
Affiliation(s)
- Christy M. Kelley
- Department of Neurobiology, Barrow Neurological Institute, Phoenix, AZ, USA
| | - Stephen D. Ginsberg
- Center for Dementia Research, Nathan Kline Institute, Orangeburg, NY, USA
- Department of Psychiatry, NYU Langone School of Medicine, New York, NY, USA
- Department of Neuroscience & Physiology, NYU Langone School of Medicine, New York, NY, USA
- NYU Neuroscience Institute, NYU Langone School of Medicine, New York, NY, USA
| | - Melissa J. Alldred
- Center for Dementia Research, Nathan Kline Institute, Orangeburg, NY, USA
- Department of Psychiatry, NYU Langone School of Medicine, New York, NY, USA
| | - Barbara J. Strupp
- Division of Nutritional Sciences and Department of Psychology, Cornell University, Ithaca, NY, USA
| | - Elliott J. Mufson
- Department of Neurobiology, Barrow Neurological Institute, Phoenix, AZ, USA
| |
Collapse
|
26
|
Zhao L, Yuan X, Wang J, Feng Y, Ji F, Li Z, Bian J. A review on flavones targeting serine/threonine protein kinases for potential anticancer drugs. Bioorg Med Chem 2019; 27:677-685. [DOI: 10.1016/j.bmc.2019.01.027] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Revised: 01/10/2019] [Accepted: 01/16/2019] [Indexed: 02/07/2023]
|
27
|
Chen R, Zhu M, Chaudhari RR, Robles O, Chen Y, Skillern W, Qin Q, Wierda WG, Zhang S, Hull KG, Romo D, Plunkett W. Creating novel translation inhibitors to target pro-survival proteins in chronic lymphocytic leukemia. Leukemia 2019; 33:1663-1674. [DOI: 10.1038/s41375-018-0364-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Revised: 10/30/2018] [Accepted: 12/03/2018] [Indexed: 12/20/2022]
|
28
|
Litzow MR, Wang XV, Carroll MP, Karp JE, Ketterling RP, Zhang Y, Kaufmann SH, Lazarus HM, Luger SM, Paietta EM, Pratz KW, Tun HW, Altman JK, Broun ER, Rybka WB, Rowe JM, Tallman MS. A randomized trial of three novel regimens for recurrent acute myeloid leukemia demonstrates the continuing challenge of treating this difficult disease. Am J Hematol 2019; 94:111-117. [PMID: 30370956 DOI: 10.1002/ajh.25333] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 10/21/2018] [Accepted: 10/24/2018] [Indexed: 12/19/2022]
Abstract
To improve the outcome of relapsed/refractory acute myeloid leukemia (AML), a randomized phase II trial of three novel regimens was conducted. Ninety patients were enrolled and were in first relapse or were refractory to induction/re-induction chemotherapy. They were randomized to the following regimens: carboplatin-topotecan (CT), each by continuous infusion for 5 days; alvocidib (formerly flavopiridol), cytarabine, and mitoxantrone (FLAM) in a timed sequential regimen; or sirolimus combined with mitoxantrone, etoposide, and cytarabine (S-MEC). The primary objective was attainment of a complete remission (CR). A Simon two-stage design was used for each of the three arms. The median age of the patients in the FLAM arm was older at 62 years compared with 55 years for the CT arm and the S-MEC arm. The overall response was 14% in the CT arm (5/35, 90% CI 7%-35%), 28% in the FLAM arm (10/36, 90% CI, 16%-43%), and 16% in the S-MEC arm (3/19, 90% CI, 4%-36%). There were nine treatment-related deaths, seven of which occurred in the FLAM arm with four of these in elderly patients. We conclude that the FLAM regimen had an encouraging response rate and should be considered for further clinical development but should be used with caution in elderly patients.
Collapse
Affiliation(s)
- Mark R. Litzow
- Departments of Hematology; Mayo Clinic; Rochester Minnesota
| | - Xin V. Wang
- Biostatistics and Computational Biology; Dana Farber Cancer Institute; Boston Massachusetts
| | - Martin P. Carroll
- Oncology; Hospital of the University of Pennsylvania; Philadelphia Pennsylvania
| | - Judith E. Karp
- Hematology/Medical Oncology; Johns Hopkins University; Baltimore Maryland
| | - Rhett P. Ketterling
- Departments of Laboratory Medicine and Pathology; Mayo Clinic; Rochester Minnesota
| | - Yanming Zhang
- Departments of Pathology; Memorial Sloan Kettering Cancer Center; New York New York
| | | | - Hillard M. Lazarus
- Seidman Cancer Center; University Hospitals Cleveland Medical Center, Case Western Reserve University; Cleveland Ohio
| | - Selina M. Luger
- Oncology; Hospital of the University of Pennsylvania; Philadelphia Pennsylvania
| | - Elisabeth M. Paietta
- Oncology; Albert Einstein College of Medicine, Montefiore Medical Center; Bronx New York
| | - Keith W. Pratz
- Hematology/Medical Oncology; Johns Hopkins University; Baltimore Maryland
| | - Han Win Tun
- Hematology/Oncology; Mayo Clinic; Jacksonville Florida
| | - Jessica K. Altman
- Hematology/Medical Oncology; Northwestern University School of Medicine; Chicago Illinois
| | - Edward R. Broun
- Hematology/Oncology; Oncology Hematology Care, Inc, Jewish Hospital; Cincinnati Ohio
| | - Witold B. Rybka
- Medicine and Pathology; Penn State Hershey Cancer Institute; Hershey Pennsylvania
| | - Jacob M. Rowe
- Department of Hematology; Shaare Zedek Medical Center; Jerusalem Israel
| | - Martin S. Tallman
- Hematology/Oncology; Memorial Sloan Kettering Cancer Center; New York New York
| |
Collapse
|
29
|
Lloret-Llinares M, Karadoulama E, Chen Y, Wojenski LA, Villafano GJ, Bornholdt J, Andersson R, Core L, Sandelin A, Jensen TH. The RNA exosome contributes to gene expression regulation during stem cell differentiation. Nucleic Acids Res 2018; 46:11502-11513. [PMID: 30212902 PMCID: PMC6265456 DOI: 10.1093/nar/gky817] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Revised: 08/29/2018] [Accepted: 09/09/2018] [Indexed: 12/27/2022] Open
Abstract
Gene expression programs change during cellular transitions. It is well established that a network of transcription factors and chromatin modifiers regulate RNA levels during embryonic stem cell (ESC) differentiation, but the full impact of post-transcriptional processes remains elusive. While cytoplasmic RNA turnover mechanisms have been implicated in differentiation, the contribution of nuclear RNA decay has not been investigated. Here, we differentiate mouse ESCs, depleted for the ribonucleolytic RNA exosome, into embryoid bodies to determine to which degree RNA abundance in the two states can be attributed to changes in transcription versus RNA decay by the exosome. As a general observation, we find that exosome depletion mainly leads to the stabilization of RNAs from lowly transcribed loci, including several protein-coding genes. Depletion of the nuclear exosome cofactor RBM7 leads to similar effects. In particular, transcripts that are differentially expressed between states tend to be more exosome sensitive in the state where expression is low. We conclude that the RNA exosome contributes to down-regulation of transcripts with disparate expression, often in conjunction with transcriptional down-regulation.
Collapse
Affiliation(s)
| | - Evdoxia Karadoulama
- Department of Molecular Biology and Genetics, Aarhus University, Denmark
- The Bioinformatics Centre, Department of Biology, University of Copenhagen, Denmark
- Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Denmark
| | - Yun Chen
- The Bioinformatics Centre, Department of Biology, University of Copenhagen, Denmark
- Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Denmark
| | - Luke A Wojenski
- Department of Molecular and Cell Biology, Institute for Systems Genomics, University of Connecticut, Storrs, CT, USA
| | - Geno J Villafano
- Department of Molecular and Cell Biology, Institute for Systems Genomics, University of Connecticut, Storrs, CT, USA
| | - Jette Bornholdt
- The Bioinformatics Centre, Department of Biology, University of Copenhagen, Denmark
- Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Denmark
| | - Robin Andersson
- The Bioinformatics Centre, Department of Biology, University of Copenhagen, Denmark
| | - Leighton Core
- Department of Molecular and Cell Biology, Institute for Systems Genomics, University of Connecticut, Storrs, CT, USA
| | - Albin Sandelin
- The Bioinformatics Centre, Department of Biology, University of Copenhagen, Denmark
- Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Denmark
| | | |
Collapse
|
30
|
Impact of elevated anti-apoptotic MCL-1 and BCL-2 on the development and treatment of MLL-AF9 AML in mice. Cell Death Differ 2018; 26:1316-1331. [PMID: 30470795 PMCID: PMC6748137 DOI: 10.1038/s41418-018-0209-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Revised: 09/19/2018] [Accepted: 09/20/2018] [Indexed: 12/13/2022] Open
Abstract
Many acute myeloid leukaemias (AMLs) express high levels of BCL-2 and MCL-1, especially after therapy. To test the impact of these anti-apoptotic proteins on AML development and treatment, we used haemopoietic reconstitution to generate MLL-AF9 AMLs expressing BCL-2 or Mcl-1 transgenes. AMLs with elevated BCL-2 or MCL-1 had a higher proportion of mature myeloid cells but, like conventional MLL-AF9 AMLs, were readily transplantable. Short-term cell lines established from multiple primary AMLs of each genotype were tested in vitro for susceptibility to chemotherapeutics currently used for treating AML (daunorubicin, etoposide, cytarabine); the proteasome inhibitor bortezomib; CDK7/9 inhibitors; and BH3 mimetics, which bind and inhibit pro-survival proteins. The BH3 mimetics tested, alone and in combination with the other drugs, were: ABT-737 which, like its clinical counterpart navitoclax, targets BCL-2, BCL-XL and BCL-W; BCL-2-specific ABT-199 (venetoclax); BCL-XL-specific A-1331852; and S63845, a new MCL-1-specific BH3 mimetic. As single agents, daunorubicin and bortezomib had the greatest efficacy. Elevated MCL-1 or BCL-2 reduced sensitivity to daunorubicin but, surprisingly, not to bortezomib. MCL-1 markedly enhanced resistance to ABT-737 and ABT-199 but not S63845, and BCL-2 increased resistance to S63845 but not to ABT-737 or ABT-199. Notable synergies were achieved by combining BH3 mimetics with daunorubicin: S63845 increased the sensitivity of both MCL-1 and BCL-2 overexpressing MLL-AF9 AMLs, and ABT-737 aided in killing those overexpressing BCL-2. Synergy between daunorubicin and ABT-199 was also apparent in vivo, although not curative. Impressive synergistic responses were achieved for human MLL-fusion AML cell lines treated with daunorubicin plus either ABT-737, ABT-199 or S63845, and with ABT-199 plus S63845, with or without daunorubicin. Our data suggest that AML patients may benefit from combining conventional cytotoxic drugs with BH3 mimetics targeting BCL-2 or MCL-1 or, if tolerated, both these agents.
Collapse
|
31
|
Dogar AM, Pauchard-Batschulat R, Grisoni-Neupert B, Richman L, Paillusson A, Pradervand S, Hagenbüchle O, Ambrosini G, Schmid CD, Bucher P, Kühn LC. Short-lived AUF1 p42-binding mRNAs of RANKL and BCL6 have two distinct instability elements each. PLoS One 2018; 13:e0206823. [PMID: 30418981 PMCID: PMC6231638 DOI: 10.1371/journal.pone.0206823] [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: 08/05/2018] [Accepted: 10/20/2018] [Indexed: 11/19/2022] Open
Abstract
Regulation of mRNA stability by RNA-protein interactions contributes significantly to quantitative aspects of gene expression. We have identified potential mRNA targets of the AU-rich element binding protein AUF1. Myc-tagged AUF1 p42 was induced in mouse NIH/3T3 cells and RNA-protein complexes isolated using anti-myc tag antibody beads. Bound mRNAs were analyzed with Affymetrix microarrays. We have identified 508 potential target mRNAs that were at least 3-fold enriched compared to control cells without myc-AUF1. 22.3% of the enriched mRNAs had an AU-rich cluster in the ARED Organism database, against 16.3% of non-enriched control mRNAs. The enrichment towards AU-rich elements was also visible by AREScore with an average value of 5.2 in the enriched mRNAs versus 4.2 in the control group. Yet, numerous mRNAs were enriched without a high ARE score. The enrichment of tetrameric and pentameric sequences suggests a broad AUF1 p42-binding spectrum at short U-rich sequences flanked by A or G. Still, some enriched mRNAs were highly unstable, as those of TNFSF11 (known as RANKL), KLF10, HES1, CCNT2, SMAD6, and BCL6. We have mapped some of the instability determinants. HES1 mRNA appeared to have a coding region determinant. Detailed analysis of the RANKL and BCL6 3’UTR revealed for both that full instability required two elements, which are conserved in evolution. In RANKL mRNA both elements are AU-rich and separated by 30 bases, while in BCL6 mRNA one is AU-rich and 60 bases from a non AU-rich element that potentially forms a stem-loop structure.
Collapse
Affiliation(s)
- Afzal M. Dogar
- Ecole Polytechnique Fédérale de Lausanne (EPFL), SV—Sciences de la Vie, ISREC—Swiss Institute for Experimental Cancer Research, Lausanne, Switzerland
| | - Ramona Pauchard-Batschulat
- Ecole Polytechnique Fédérale de Lausanne (EPFL), SV—Sciences de la Vie, ISREC—Swiss Institute for Experimental Cancer Research, Lausanne, Switzerland
| | - Barbara Grisoni-Neupert
- Ecole Polytechnique Fédérale de Lausanne (EPFL), SV—Sciences de la Vie, ISREC—Swiss Institute for Experimental Cancer Research, Lausanne, Switzerland
| | - Larry Richman
- Ecole Polytechnique Fédérale de Lausanne (EPFL), SV—Sciences de la Vie, ISREC—Swiss Institute for Experimental Cancer Research, Lausanne, Switzerland
| | - Alexandra Paillusson
- Center for Integrative Genomics (CIG), University of Lausanne, Génopode, Lausanne, Switzerland
| | - Sylvain Pradervand
- Center for Integrative Genomics (CIG), University of Lausanne, Génopode, Lausanne, Switzerland
| | - Otto Hagenbüchle
- Ecole Polytechnique Fédérale de Lausanne (EPFL), SV—Sciences de la Vie, ISREC—Swiss Institute for Experimental Cancer Research, Lausanne, Switzerland
- Center for Integrative Genomics (CIG), University of Lausanne, Génopode, Lausanne, Switzerland
| | - Giovanna Ambrosini
- Ecole Polytechnique Fédérale de Lausanne (EPFL), SV—Sciences de la Vie, ISREC—Swiss Institute for Experimental Cancer Research, Lausanne, Switzerland
- Swiss Institute of Bioinformatics (SIB), Lausanne, Switzerland
| | | | - Philipp Bucher
- Ecole Polytechnique Fédérale de Lausanne (EPFL), SV—Sciences de la Vie, ISREC—Swiss Institute for Experimental Cancer Research, Lausanne, Switzerland
- Swiss Institute of Bioinformatics (SIB), Lausanne, Switzerland
| | - Lukas C. Kühn
- Ecole Polytechnique Fédérale de Lausanne (EPFL), SV—Sciences de la Vie, ISREC—Swiss Institute for Experimental Cancer Research, Lausanne, Switzerland
- * E-mail:
| |
Collapse
|
32
|
Abstract
The fact that many cancer types display transcriptional addiction driven by dysregulation of oncogenic enhancers and transcription factors has led to increased interest in a group of protein kinases, known as transcriptional cyclin dependent kinases (tCDKs), as potential therapeutic targets. Despite early reservations about targeting a process that is essential to healthy cell types, there is now evidence that targeting tCDKs could provide enough therapeutic window to be effective in the clinic. Here, we discuss recent developments in this field, with an emphasis on highly-selective inhibitors and the challenges to be addressed before these inhibitors could be used for therapeutic purposes. Abbreviations: CAK: CDK-activating kinase;CDK: cyclin-dependent kinase;CMGC group: CDK-, MAPK-, GSK3-, and CLK-like;CTD: C-terminal repeat domain of the RPB1 subunit of RNA polymerase II;DRB: 5,6-dichloro-1-β-D-ribofuranosylbenzimidazole;mCRPC: metastatic castration-resistant prostate cancer;NSCLC: non-small cell lung cancer;P-TEFb: positive elongation factor b;RNAPII: RNA polymerase II;S2: serine-2 of CTD repeats;S5: serine-5 of CTD repeats;S7: serine-7 of CTD repeats;SEC: super elongation complex;tCDK: transcriptional cyclin-dependent kinase;TNBC: triple-negative breast cancer
Collapse
Affiliation(s)
- Matthew D Galbraith
- a Linda Crnic Institute for Down Syndrome, School of Medicine , University of Colorado Anschutz Medical Campus , Aurora , CO , USA.,b Department of Pharmacology, School of Medicine , University of Colorado Anschutz Medical Campus , Aurora , CO , USA
| | - Heather Bender
- a Linda Crnic Institute for Down Syndrome, School of Medicine , University of Colorado Anschutz Medical Campus , Aurora , CO , USA.,b Department of Pharmacology, School of Medicine , University of Colorado Anschutz Medical Campus , Aurora , CO , USA
| | - Joaquín M Espinosa
- a Linda Crnic Institute for Down Syndrome, School of Medicine , University of Colorado Anschutz Medical Campus , Aurora , CO , USA.,b Department of Pharmacology, School of Medicine , University of Colorado Anschutz Medical Campus , Aurora , CO , USA.,c Department of Molecular, Cellular and Developmental Biology , University of Colorado Boulder , Boulder , CO , USA
| |
Collapse
|
33
|
Göthert JR, Imsak R, Möllmann M, Kesper S, Göbel M, Dührsen U, Scholz A, Lücking U, Baumann M, Unger A, Schultz-Fademrecht C, Klebl B, Eickhoff J, Choidas A, Dürig J. Potent anti-leukemic activity of a specific cyclin-dependent kinase 9 inhibitor in mouse models of chronic lymphocytic leukemia. Oncotarget 2018; 9:26353-26369. [PMID: 29899864 PMCID: PMC5995184 DOI: 10.18632/oncotarget.25293] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Accepted: 04/07/2018] [Indexed: 12/23/2022] Open
Abstract
Onset of progression even during therapy with novel drugs remains an issue in chronic lymphocytic leukemia (CLL). Thus, there is ongoing demand for novel agents. Approaches targeting cyclin-dependent kinases (CDK) have reached the clinical trial stage. CDK9 mediating RNA transcriptional elongation is the evolving pivotal CLL CDK inhibitor target. However, more CDK9 selective compounds are desirable. Here, we describe the CDK9 inhibitor LDC526 displaying a low nanomolar biochemical activity against CDK9 and an at least 50-fold selectivity against other CDKs. After demonstrating in vitro MEC-1 cell line and primary human CLL cell cytotoxicity we evaluated the LDC526 in vivo effect on human CLL cells transplanted into NOD/scid/γcnull (NSG) mice. LDC526 administration (75 mg/kg) for 5 days resulted in a 77% reduction of human CLL cells in NSG spleens compared to carrier control treatment. Next, we longitudinally studied the LDC526 impact on circulating CLL cells in the TCL1 transgenic mouse model. LDC526 (50 mg/kg) administration for two days led to a 16-fold reduction of blood CLL cell numbers. Remarkably, residual CLL cells exhibited significantly increased intracellular BCL-2 levels. However, the LDC526 cytotoxic effect was not restricted to CLL cells as also declining numbers of normal B and T lymphocytes were observed in LDC526 treated TCL1 mice. Taken together, our in vivo data provide a strong rational for continued LDC526 development in CLL therapy and argue for the combination with BCL-2 inhibitors.
Collapse
Affiliation(s)
- Joachim R Göthert
- Department of Hematology, West German Cancer Center (WTZ), University Hospital Essen, Essen, Germany
| | - Roze Imsak
- Department of Hematology, West German Cancer Center (WTZ), University Hospital Essen, Essen, Germany
| | - Michael Möllmann
- Department of Hematology, West German Cancer Center (WTZ), University Hospital Essen, Essen, Germany
| | - Stefanie Kesper
- Department of Hematology, West German Cancer Center (WTZ), University Hospital Essen, Essen, Germany
| | - Maria Göbel
- Department of Hematology, West German Cancer Center (WTZ), University Hospital Essen, Essen, Germany
| | - Ulrich Dührsen
- Department of Hematology, West German Cancer Center (WTZ), University Hospital Essen, Essen, Germany
| | - Arne Scholz
- Bayer AG, Pharmaceuticals, Drug Discovery, Berlin, Germany
| | - Ulrich Lücking
- Bayer AG, Pharmaceuticals, Drug Discovery, Berlin, Germany
| | | | - Anke Unger
- Lead Discovery Center GmbH (LDC), Dortmund, Germany
| | | | - Bert Klebl
- Lead Discovery Center GmbH (LDC), Dortmund, Germany
| | - Jan Eickhoff
- Lead Discovery Center GmbH (LDC), Dortmund, Germany
| | - Axel Choidas
- Lead Discovery Center GmbH (LDC), Dortmund, Germany
| | - Jan Dürig
- Department of Hematology, West German Cancer Center (WTZ), University Hospital Essen, Essen, Germany
| |
Collapse
|
34
|
Targeting General Transcriptional Machinery as a Therapeutic Strategy for Adult T-Cell Leukemia. Molecules 2018; 23:molecules23051057. [PMID: 29724031 PMCID: PMC6099935 DOI: 10.3390/molecules23051057] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2018] [Revised: 04/27/2018] [Accepted: 04/28/2018] [Indexed: 12/18/2022] Open
Abstract
Cancer cells are highly reliant on certain molecular pathways, which support their survival and proliferation. The fundamental concept of molecularly targeted therapy is to target a protein that is specifically deregulated or overexpressed in cancer cells. However, drug resistance and tumor heterogeneity are major obstacles in the development of specific inhibitors. Additionally, many driver oncogenes exert their oncogenic property via abnormal expression without having genetic mutations. Interestingly, recent accumulating evidence has demonstrated that many critical cancer genes are driven by a unique class of enhancers termed super-enhancers. Genes associated with super-enhancers are relatively more susceptible to the inhibition of general transcriptional machinery compared with genes that are regulated by typical enhancers. Cancer cells are more sensitive to treatment with small-molecule inhibitors of CDK7 or BRD4 than non-transformed cells. These findings proposed a novel strategy to identify functionally important genes as well as novel therapeutic modalities in cancer. This approach would be particularly useful for genetically complicated cancers, such as adult T-cell leukemia (ATL), whereby a large mutational burden is present, but the functional consequences of each mutation have not been well-studied. In this review, we discuss recent findings on super-enhancers, underlying mechanisms, and the efficacy of small-molecule transcriptional inhibitors in ATL.
Collapse
|
35
|
Schrecengost RS, Green CL, Zhuang Y, Keller SN, Smith RA, Maines LW, Smith CD. In Vitro and In Vivo Antitumor and Anti-Inflammatory Capabilities of the Novel GSK3 and CDK9 Inhibitor ABC1183. J Pharmacol Exp Ther 2018; 365:107-116. [PMID: 29434052 PMCID: PMC5830635 DOI: 10.1124/jpet.117.245738] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Accepted: 01/19/2018] [Indexed: 01/20/2023] Open
Abstract
Glycogen synthase kinase-3s (GSK3α and GSK3β) are constitutively active protein kinases that target over 100 substrates, incorporate into numerous protein complexes, and regulate such vital cellular functions as proliferation, apoptosis, and inflammation. Cyclin-dependent kinase 9 (CDK9) regulates RNA production as a component of positive transcription elongation factor b and promotes expression of oncogenic and inflammatory genes. Simultaneous inhibition of these signaling nodes is a promising approach for drug discovery, although previous compounds exhibit limited selectivity and clinical efficacy. The novel diaminothiazole ABC1183 is a selective GSK3α/β and CDK9 inhibitor and is growth-inhibitory against a broad panel of cancer cell lines. ABC1183 treatment decreases cell survival through G2/M arrest and modulates oncogenic signaling through changes in GSK3, glycogen synthase, and β-catenin phosphorylation and MCL1 expression. Oral administration, which demonstrates no organ or hematologic toxicity, suppresses tumor growth and inflammation-driven gastrointestinal disease symptoms, owing in part to downregulation of tumor necrosis factor α and interleukin-6 proinflammatory cytokines. Therefore, ABC1183 is strategically poised to effectively mitigate multiple clinically relevant diseases.
Collapse
Affiliation(s)
| | | | - Yan Zhuang
- Apogee Biotechnology Corporation, Hummelstown, Pennsylvania
| | - Staci N Keller
- Apogee Biotechnology Corporation, Hummelstown, Pennsylvania
| | - Ryan A Smith
- Apogee Biotechnology Corporation, Hummelstown, Pennsylvania
| | - Lynn W Maines
- Apogee Biotechnology Corporation, Hummelstown, Pennsylvania
| | | |
Collapse
|
36
|
Whittaker SR, Barlow C, Martin MP, Mancusi C, Wagner S, Self A, Barrie E, Te Poele R, Sharp S, Brown N, Wilson S, Jackson W, Fischer PM, Clarke PA, Walton MI, McDonald E, Blagg J, Noble M, Garrett MD, Workman P. Molecular profiling and combinatorial activity of CCT068127: a potent CDK2 and CDK9 inhibitor. Mol Oncol 2018; 12:287-304. [PMID: 29063678 PMCID: PMC5830651 DOI: 10.1002/1878-0261.12148] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Revised: 10/05/2017] [Accepted: 10/07/2017] [Indexed: 01/18/2023] Open
Abstract
Deregulation of the cyclin-dependent kinases (CDKs) has been implicated in the pathogenesis of multiple cancer types. Consequently, CDKs have garnered intense interest as therapeutic targets for the treatment of cancer. We describe herein the molecular and cellular effects of CCT068127, a novel inhibitor of CDK2 and CDK9. Optimized from the purine template of seliciclib, CCT068127 exhibits greater potency and selectivity against purified CDK2 and CDK9 and superior antiproliferative activity against human colon cancer and melanoma cell lines. X-ray crystallography studies reveal that hydrogen bonding with the DFG motif of CDK2 is the likely mechanism of greater enzymatic potency. Commensurate with inhibition of CDK activity, CCT068127 treatment results in decreased retinoblastoma protein (RB) phosphorylation, reduced phosphorylation of RNA polymerase II, and induction of cell cycle arrest and apoptosis. The transcriptional signature of CCT068127 shows greatest similarity to other small-molecule CDK and also HDAC inhibitors. CCT068127 caused a dramatic loss in expression of DUSP6 phosphatase, alongside elevated ERK phosphorylation and activation of MAPK pathway target genes. MCL1 protein levels are rapidly decreased by CCT068127 treatment and this associates with synergistic antiproliferative activity after combined treatment with CCT068127 and ABT263, a BCL2 family inhibitor. These findings support the rational combination of this series of CDK2/9 inhibitors and BCL2 family inhibitors for the treatment of human cancer.
Collapse
Affiliation(s)
- Steven R. Whittaker
- Cancer Research UK Cancer Therapeutics UnitDivision of Cancer TherapeuticsThe Institute of Cancer ResearchLondonUK
| | - Clare Barlow
- Cancer Research UK Cancer Therapeutics UnitDivision of Cancer TherapeuticsThe Institute of Cancer ResearchLondonUK
| | - Mathew P. Martin
- Northern Institute for Cancer ResearchUniversity of Newcastle upon TyneMedical SchoolNewcastle upon TyneUK
| | - Caterina Mancusi
- Cancer Research UK Cancer Therapeutics UnitDivision of Cancer TherapeuticsThe Institute of Cancer ResearchLondonUK
| | - Steve Wagner
- Cancer Research UK Cancer Therapeutics UnitDivision of Cancer TherapeuticsThe Institute of Cancer ResearchLondonUK
| | - Annette Self
- Cancer Research UK Cancer Therapeutics UnitDivision of Cancer TherapeuticsThe Institute of Cancer ResearchLondonUK
| | - Elaine Barrie
- Cancer Research UK Cancer Therapeutics UnitDivision of Cancer TherapeuticsThe Institute of Cancer ResearchLondonUK
| | - Robert Te Poele
- Cancer Research UK Cancer Therapeutics UnitDivision of Cancer TherapeuticsThe Institute of Cancer ResearchLondonUK
| | - Swee Sharp
- Cancer Research UK Cancer Therapeutics UnitDivision of Cancer TherapeuticsThe Institute of Cancer ResearchLondonUK
| | - Nathan Brown
- Cancer Research UK Cancer Therapeutics UnitDivision of Cancer TherapeuticsThe Institute of Cancer ResearchLondonUK
| | - Stuart Wilson
- Cancer Research UK Cancer Therapeutics UnitDivision of Cancer TherapeuticsThe Institute of Cancer ResearchLondonUK
| | - Wayne Jackson
- Cyclacel Ltd.DundeeUK
- Present address:
Samuel Lister AcademyBingleyWest YorkshireBD16 1TZUK
| | - Peter M. Fischer
- Cyclacel Ltd.DundeeUK
- Present address:
School of Pharmacy and Centre for Biomolecular SciencesUniversity of Nottingham, University ParkNottinghamNG7 2RDUK
| | - Paul A. Clarke
- Cancer Research UK Cancer Therapeutics UnitDivision of Cancer TherapeuticsThe Institute of Cancer ResearchLondonUK
| | - Michael I. Walton
- Cancer Research UK Cancer Therapeutics UnitDivision of Cancer TherapeuticsThe Institute of Cancer ResearchLondonUK
| | - Edward McDonald
- Cancer Research UK Cancer Therapeutics UnitDivision of Cancer TherapeuticsThe Institute of Cancer ResearchLondonUK
| | - Julian Blagg
- Cancer Research UK Cancer Therapeutics UnitDivision of Cancer TherapeuticsThe Institute of Cancer ResearchLondonUK
| | - Martin Noble
- Northern Institute for Cancer ResearchUniversity of Newcastle upon TyneMedical SchoolNewcastle upon TyneUK
| | - Michelle D. Garrett
- Cancer Research UK Cancer Therapeutics UnitDivision of Cancer TherapeuticsThe Institute of Cancer ResearchLondonUK
- Present address:
School of BiosciencesUniversity of KentCanterburyKentCT2 7NJUK
| | - Paul Workman
- Cancer Research UK Cancer Therapeutics UnitDivision of Cancer TherapeuticsThe Institute of Cancer ResearchLondonUK
| |
Collapse
|
37
|
Lugowski A, Nicholson B, Rissland OS. Determining mRNA half-lives on a transcriptome-wide scale. Methods 2018; 137:90-98. [DOI: 10.1016/j.ymeth.2017.12.006] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Revised: 12/11/2017] [Accepted: 12/11/2017] [Indexed: 12/20/2022] Open
|
38
|
Su YT, Chen R, Wang H, Song H, Zhang Q, Chen LY, Lappin H, Vasconcelos G, Lita A, Maric D, Li A, Celiku O, Zhang W, Meetze K, Estok T, Larion M, Abu-Asab M, Zhuang Z, Yang C, Gilbert MR, Wu J. Novel Targeting of Transcription and Metabolism in Glioblastoma. Clin Cancer Res 2017; 24:1124-1137. [PMID: 29254993 DOI: 10.1158/1078-0432.ccr-17-2032] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Revised: 10/31/2017] [Accepted: 12/13/2017] [Indexed: 11/16/2022]
Abstract
Purpose: Glioblastoma (GBM) is highly resistant to treatment, largely due to disease heterogeneity and resistance mechanisms. We sought to investigate a promising drug that can inhibit multiple aspects of cancer cell survival mechanisms and become an effective therapeutic for GBM patients.Experimental Design: To investigate TG02, an agent with known penetration of the blood-brain barrier, we examined the effects as single agent and in combination with temozolomide, a commonly used chemotherapy in GBM. We used human GBM cells and a syngeneic mouse orthotopic GBM model, evaluating survival and the pharmacodynamics of TG02. Mechanistic studies included TG02-induced transcriptional regulation, apoptosis, and RNA sequencing in treated GBM cells as well as the investigation of mitochondrial and glycolytic function assays.Results: We demonstrated that TG02 inhibited cell proliferation, induced cell death, and synergized with temozolomide in GBM cells with different genetic background but not in astrocytes. TG02-induced cytotoxicity was blocked by the overexpression of phosphorylated CDK9, suggesting a CDK9-dependent cell killing. TG02 suppressed transcriptional progression of antiapoptotic proteins and induced apoptosis in GBM cells. We further demonstrated that TG02 caused mitochondrial dysfunction and glycolytic suppression and ultimately ATP depletion in GBM. A prolonged survival was observed in GBM mice receiving combined treatment of TG02 and temozolomide. The TG02-induced decrease of CDK9 phosphorylation was confirmed in the brain tumor tissue.Conclusions: TG02 inhibits multiple survival mechanisms and synergistically decreases energy production with temozolomide, representing a promising therapeutic strategy in GBM, currently under investigation in an ongoing clinical trial. Clin Cancer Res; 24(5); 1124-37. ©2017 AACR.
Collapse
Affiliation(s)
- Yu-Ting Su
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Robert Chen
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Herui Wang
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Hua Song
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Qi Zhang
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Li-Yuan Chen
- Critical Care Medicine Department, Clinical Center, NIH, Bethesda, Maryland
| | - Hallie Lappin
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Gabriel Vasconcelos
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Adrian Lita
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Dragan Maric
- Flow and Imaging Cytometry Core Facility, National Institute of Neurological Disorders and Stroke, NIH, Bethesda, Maryland
| | - Aiguo Li
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Orieta Celiku
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Wei Zhang
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | | | | | - Mioara Larion
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Mones Abu-Asab
- Section of Histopathology, National Eye Institute, NIH, Bethesda, Maryland
| | - Zhengping Zhuang
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Chunzhang Yang
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Mark R Gilbert
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Jing Wu
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland.
| |
Collapse
|
39
|
Bernardini A, Martínez JL. Genome-wide analysis shows that RNase G plays a global role in the stability of mRNAs in Stenotrophomonas maltophilia. Sci Rep 2017; 7:16016. [PMID: 29167539 PMCID: PMC5700063 DOI: 10.1038/s41598-017-16091-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Accepted: 11/07/2017] [Indexed: 11/28/2022] Open
Abstract
Gene expression is determined by critical processes such as RNA synthesis and degradation. Ribonucleases participate in the coordinated and differential decay of messenger RNAs. We describe a suitable method of normalization and calculation of mRNAs half-life values quantified by RNA-Seq. We determined the mRNA half-lives of more than 2000 genes in Stenotrophomonas maltophilia D457 and in an isogenic RNase G deficient mutant. Median half-lives were 2,74 and 3 min in the wild-type and the rng-deficient strain, respectively. The absence of RNase G resulted in an overall enhancement of mRNA half-life times, showing that many RNAs are targets of RNase G in S. maltophilia. Around 40 genes are likely to be regulated directly by RNase G since their half-lives were more than two-fold higher in the rng-deficient mutant. Gene length, GC content or expression levels did not correlate with mRNAs lifetimes, although groups of genes with different functions showed different RNA half-lives. Further, we predicted 1542 gene pairs to be part of the same operons in S. maltophilia. In contrast to what was described for other bacteria, our data indicate that RNase G has a global role in mRNA stability and consequently in the regulation of S. maltophilia gene expression.
Collapse
Affiliation(s)
| | - José L Martínez
- Centro Nacional de Biotecnología, CSIC, Darwin 3, 28049, Madrid, Spain.
| |
Collapse
|
40
|
Johnson SF, Cruz C, Greifenberg AK, Dust S, Stover DG, Chi D, Primack B, Cao S, Bernhardy AJ, Coulson R, Lazaro JB, Kochupurakkal B, Sun H, Unitt C, Moreau LA, Sarosiek KA, Scaltriti M, Juric D, Baselga J, Richardson AL, Rodig SJ, D'Andrea AD, Balmaña J, Johnson N, Geyer M, Serra V, Lim E, Shapiro GI. CDK12 Inhibition Reverses De Novo and Acquired PARP Inhibitor Resistance in BRCA Wild-Type and Mutated Models of Triple-Negative Breast Cancer. Cell Rep 2017; 17:2367-2381. [PMID: 27880910 DOI: 10.1016/j.celrep.2016.10.077] [Citation(s) in RCA: 209] [Impact Index Per Article: 26.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Revised: 09/08/2016] [Accepted: 10/23/2016] [Indexed: 02/04/2023] Open
Abstract
Although poly(ADP-ribose) polymerase (PARP) inhibitors are active in homologous recombination (HR)-deficient cancers, their utility is limited by acquired resistance after restoration of HR. Here, we report that dinaciclib, an inhibitor of cyclin-dependent kinases (CDKs) 1, 2, 5, and 9, additionally has potent activity against CDK12, a transcriptional regulator of HR. In BRCA-mutated triple-negative breast cancer (TNBC) cells and patient-derived xenografts (PDXs), dinaciclib ablates restored HR and reverses PARP inhibitor resistance. Additionally, we show that de novo resistance to PARP inhibition in BRCA1-mutated cell lines and a PDX derived from a PARP-inhibitor-naive BRCA1 carrier is mediated by residual HR and is reversed by CDK12 inhibition. Finally, dinaciclib augments the degree of response in a PARP-inhibitor-sensitive model, converting tumor growth inhibition to durable regression. These results highlight the significance of HR disruption as a therapeutic strategy and support the broad use of combined CDK12 and PARP inhibition in TNBC.
Collapse
Affiliation(s)
- Shawn F Johnson
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Cristina Cruz
- Experimental Therapeutics Group, Vall d'Hebron Institute of Oncology, 08035 Barcelona, Spain; Medical Oncology Department, Hospital Vall d'Hebron, Vall d'Hebron Institute of Oncology, Universitat Autonoma de Barcelona, 08035 Barcelona, Spain
| | - Ann Katrin Greifenberg
- Department of Structural Immunology, Institute of Innate Immunity, University of Bonn, 53127 Bonn, Germany; Group Physical Biochemistry, Center of Advanced European Studies and Research, 53175 Bonn, Germany
| | - Sofia Dust
- Department of Structural Immunology, Institute of Innate Immunity, University of Bonn, 53127 Bonn, Germany; Group Physical Biochemistry, Center of Advanced European Studies and Research, 53175 Bonn, Germany
| | - Daniel G Stover
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA; Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - David Chi
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Benjamin Primack
- Department of Radiation Oncology and Center for DNA Damage and Repair, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA 02215, USA; Department of Pediatric Oncology, Dana-Farber Cancer Institute, Children's Hospital and Harvard Medical School, Boston, MA 02215, USA
| | - Shiliang Cao
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Andrea J Bernhardy
- Developmental Therapeutics Program, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | - Rhiannon Coulson
- The Kinghorn Cancer Centre, Garvan Institute of Medical Research, St. Vincent's Health Network, Darlinghurst, NSW 2010, Australia
| | - Jean-Bernard Lazaro
- Department of Radiation Oncology and Center for DNA Damage and Repair, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA 02215, USA
| | - Bose Kochupurakkal
- Department of Radiation Oncology and Center for DNA Damage and Repair, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA 02215, USA
| | - Heather Sun
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Christine Unitt
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Lisa A Moreau
- Department of Radiation Oncology and Center for DNA Damage and Repair, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA 02215, USA; Department of Pediatric Oncology, Dana-Farber Cancer Institute, Children's Hospital and Harvard Medical School, Boston, MA 02215, USA
| | | | - Maurizio Scaltriti
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Dejan Juric
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA 02114, USA; Department of Medicine, Harvard Medical School, Boston, MA 02114, USA
| | - José Baselga
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Andrea L Richardson
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Scott J Rodig
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Alan D D'Andrea
- Department of Radiation Oncology and Center for DNA Damage and Repair, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA 02215, USA; Department of Pediatric Oncology, Dana-Farber Cancer Institute, Children's Hospital and Harvard Medical School, Boston, MA 02215, USA
| | - Judith Balmaña
- Medical Oncology Department, Hospital Vall d'Hebron, Vall d'Hebron Institute of Oncology, Universitat Autonoma de Barcelona, 08035 Barcelona, Spain
| | - Neil Johnson
- Developmental Therapeutics Program, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | - Matthias Geyer
- Department of Structural Immunology, Institute of Innate Immunity, University of Bonn, 53127 Bonn, Germany; Group Physical Biochemistry, Center of Advanced European Studies and Research, 53175 Bonn, Germany
| | - Violeta Serra
- Experimental Therapeutics Group, Vall d'Hebron Institute of Oncology, 08035 Barcelona, Spain
| | - Elgene Lim
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; The Kinghorn Cancer Centre, Garvan Institute of Medical Research, St. Vincent's Health Network, Darlinghurst, NSW 2010, Australia.
| | - Geoffrey I Shapiro
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
41
|
Crisp PA, Ganguly DR, Smith AB, Murray KD, Estavillo GM, Searle I, Ford E, Bogdanović O, Lister R, Borevitz JO, Eichten SR, Pogson BJ. Rapid Recovery Gene Downregulation during Excess-Light Stress and Recovery in Arabidopsis. THE PLANT CELL 2017; 29:1836-1863. [PMID: 28705956 PMCID: PMC5590493 DOI: 10.1105/tpc.16.00828] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Revised: 06/22/2017] [Accepted: 07/11/2017] [Indexed: 05/19/2023]
Abstract
Stress recovery may prove to be a promising approach to increase plant performance and, theoretically, mRNA instability may facilitate faster recovery. Transcriptome (RNA-seq, qPCR, sRNA-seq, and PARE) and methylome profiling during repeated excess-light stress and recovery was performed at intervals as short as 3 min. We demonstrate that 87% of the stress-upregulated mRNAs analyzed exhibit very rapid recovery. For instance, HSP101 abundance declined 2-fold every 5.1 min. We term this phenomenon rapid recovery gene downregulation (RRGD), whereby mRNA abundance rapidly decreases promoting transcriptome resetting. Decay constants (k) were modeled using two strategies, linear and nonlinear least squares regressions, with the latter accounting for both transcription and degradation. This revealed extremely short half-lives ranging from 2.7 to 60.0 min for 222 genes. Ribosome footprinting using degradome data demonstrated RRGD loci undergo cotranslational decay and identified changes in the ribosome stalling index during stress and recovery. However, small RNAs and 5'-3' RNA decay were not essential for recovery of the transcripts examined, nor were any of the six excess light-associated methylome changes. We observed recovery-specific gene expression networks upon return to favorable conditions and six transcriptional memory types. In summary, rapid transcriptome resetting is reported in the context of active recovery and cellular memory.
Collapse
Affiliation(s)
- Peter A Crisp
- Australian Research Council Centre of Excellence in Plant Energy Biology, Research School of Biology, Australian National University Canberra, Acton ACT 0200, Australia
- Department of Plant and Microbial Biology, University of Minnesota, Saint Paul, Minnesota 55108
| | - Diep R Ganguly
- Australian Research Council Centre of Excellence in Plant Energy Biology, Research School of Biology, Australian National University Canberra, Acton ACT 0200, Australia
| | - Aaron B Smith
- Australian Research Council Centre of Excellence in Plant Energy Biology, Research School of Biology, Australian National University Canberra, Acton ACT 0200, Australia
| | - Kevin D Murray
- Australian Research Council Centre of Excellence in Plant Energy Biology, Research School of Biology, Australian National University Canberra, Acton ACT 0200, Australia
| | - Gonzalo M Estavillo
- Australian Research Council Centre of Excellence in Plant Energy Biology, Research School of Biology, Australian National University Canberra, Acton ACT 0200, Australia
- CSIRO Agriculture and Food, Black Mountain, Canberra ACT 2601, Australia
| | - Iain Searle
- School of Biological Sciences, The University of Adelaide, SA 5005, Australia
| | - Ethan Ford
- Australian Research Council Centre of Excellence in Plant Energy Biology, The University of Western Australia, Perth WA 6009, Australia
| | - Ozren Bogdanović
- Australian Research Council Centre of Excellence in Plant Energy Biology, The University of Western Australia, Perth WA 6009, Australia
- Harry Perkins Institute of Medical Research, Perth WA 6009, Australia
| | - Ryan Lister
- Australian Research Council Centre of Excellence in Plant Energy Biology, The University of Western Australia, Perth WA 6009, Australia
- Harry Perkins Institute of Medical Research, Perth WA 6009, Australia
| | - Justin O Borevitz
- Australian Research Council Centre of Excellence in Plant Energy Biology, Research School of Biology, Australian National University Canberra, Acton ACT 0200, Australia
| | - Steven R Eichten
- Australian Research Council Centre of Excellence in Plant Energy Biology, Research School of Biology, Australian National University Canberra, Acton ACT 0200, Australia
| | - Barry J Pogson
- Australian Research Council Centre of Excellence in Plant Energy Biology, Research School of Biology, Australian National University Canberra, Acton ACT 0200, Australia
| |
Collapse
|
42
|
Complementary dynamic BH3 profiles predict co-operativity between the multi-kinase inhibitor TG02 and the BH3 mimetic ABT-199 in acute myeloid leukaemia cells. Oncotarget 2017; 8:16220-16232. [PMID: 27092880 PMCID: PMC5369958 DOI: 10.18632/oncotarget.8742] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Accepted: 03/28/2016] [Indexed: 12/18/2022] Open
Abstract
Direct co-operation between sensitiser molecules BAD and NOXA in mediating apoptosis suggests that therapeutic agents which sensitise to BAD may complement agents which sensitise to NOXA. Dynamic BH3 profiling is a novel methodology that we have applied to the measurement of complementarity between sensitiser BH3 peptide mimetics and therapeutic agents. Using dynamic BH3 profiling, we show that the agent TG02, which downregulates MCL-1, sensitises to the BCL-2-inhibitory BAD-BH3 peptide, whereas the BCL-2 antagonist ABT-199 sensitises to MCL-1 inhibitory NOXA-BH3 peptide in acute myeloid leukaemia (AML) cells. At the concentrations used, the peptides did not trigger mitochondrial outer membrane permeabilisation in their own right, but primed cells to release Cytochrome C in the presence of an appropriate trigger of a complementary pathway. In KG-1a cells TG02 and ABT-199 synergised to induce apoptosis. In heterogeneous AML patient samples we noted a range of sensitivities to the two agents. Although some individual samples markedly favoured one agent or the other, in the group as a whole the combination of TG02 + ABT-199 was significantly more cytotoxic than either agent individually. We conclude that dynamic NOXA and BAD BH3 profiling is a sensitive methodology for investigating molecular pathways of drug action and complementary mechanisms of chemoresponsiveness.
Collapse
|
43
|
Abstract
Over the past two decades there has been a great deal of interest in the development of inhibitors of the cyclin-dependent kinases (CDKs). This attention initially stemmed from observations that different CDK isoforms have key roles in cancer cell proliferation through loss of regulation of the cell cycle, a hallmark feature of cancer. CDKs have now been shown to regulate other processes, particularly various aspects of transcription. The early non-selective CDK inhibitors exhibited considerable toxicity and proved to be insufficiently active in most cancers. The lack of patient selection biomarkers and an absence of understanding of the inhibitory profile required for efficacy hampered the development of these inhibitors. However, the advent of potent isoform-selective inhibitors with accompanying biomarkers has re-ignited interest. Palbociclib, a selective CDK4/6 inhibitor, is now approved for the treatment of ER+/HER2- advanced breast cancer. Current developments in the field include the identification of potent and selective inhibitors of the transcriptional CDKs; these include tool compounds that have allowed exploration of individual CDKs as cancer targets and the determination of their potential therapeutic windows. Biomarkers that allow the selection of patients likely to respond are now being discovered. Drug resistance has emerged as a major hurdle in the clinic for most protein kinase inhibitors and resistance mechanism are beginning to be identified for CDK inhibitors. This suggests that the selective inhibitors may be best used combined with standard of care or other molecularly targeted agents now in development rather than in isolation as monotherapies.
Collapse
Affiliation(s)
- Steven R Whittaker
- Division of Cancer Therapeutics, The Institute of Cancer Research, London SW7 3RP, United Kingdom
| | - Aurélie Mallinger
- Division of Cancer Therapeutics, The Institute of Cancer Research, London SW7 3RP, United Kingdom; Cancer Research UK Cancer Therapeutics Unit, The Institute of Cancer Research, London SW7 3RP, United Kingdom
| | - Paul Workman
- Division of Cancer Therapeutics, The Institute of Cancer Research, London SW7 3RP, United Kingdom; Cancer Research UK Cancer Therapeutics Unit, The Institute of Cancer Research, London SW7 3RP, United Kingdom
| | - Paul A Clarke
- Division of Cancer Therapeutics, The Institute of Cancer Research, London SW7 3RP, United Kingdom; Cancer Research UK Cancer Therapeutics Unit, The Institute of Cancer Research, London SW7 3RP, United Kingdom.
| |
Collapse
|
44
|
Matrone G, Mullins JJ, Tucker CS, Denvir MA. Effects of Cyclin Dependent Kinase 9 inhibition on zebrafish larvae. Cell Cycle 2016; 15:3060-3069. [PMID: 27715402 PMCID: PMC5134698 DOI: 10.1080/15384101.2016.1231283] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
CDK9 is a known regulator of cellular transcription, growth and proliferation. Small molecule inhibitors are currently being developed and assessed in clinical trials as anti-cancer drugs. The zebrafish embryo provides an ideal model to explore the effects of CDK9 inhibition in-vivo. This has not been adequately explored previously at the level of a whole organism. We have compared and contrasted the effects of pharmacological and molecular inhibition of CDK9 on somatic growth, apoptosis and cellular proliferation in zebrafish larvae between 0 to 120 hours post fertilisation (hpf) using flavopiridol, a selective CDK9 antagonist, and CDK9-targeting morpholino. We demonstrate that the inhibition of CDK9 diminishes cellular proliferation and increases apoptosis. Subsequently, it affects somatic growth and development of a number of key embryonic structures including the brain, heart, eye and blood vessels. For the first time, we have localized CDK9 at a subcellular level in whole-mounted larvae. This works shows, at a high-throughput level, that CDK9 clearly plays a fundamental role in early cellular growth and proliferation.
Collapse
Affiliation(s)
- Gianfranco Matrone
- a British Heart Foundation Centre for Cardiovascular Science, The Queen's Medical Research Institute, The University of Edinburgh , Edinburgh , UK.,b Department of Cardiovascular Sciences , Methodist Hospital Research Institute , Houston , TX , USA
| | - John J Mullins
- a British Heart Foundation Centre for Cardiovascular Science, The Queen's Medical Research Institute, The University of Edinburgh , Edinburgh , UK
| | - Carl S Tucker
- a British Heart Foundation Centre for Cardiovascular Science, The Queen's Medical Research Institute, The University of Edinburgh , Edinburgh , UK
| | - Martin A Denvir
- a British Heart Foundation Centre for Cardiovascular Science, The Queen's Medical Research Institute, The University of Edinburgh , Edinburgh , UK
| |
Collapse
|
45
|
Rajput S, Khera N, Guo Z, Hoog J, Li S, Ma CX. Inhibition of cyclin dependent kinase 9 by dinaciclib suppresses cyclin B1 expression and tumor growth in triple negative breast cancer. Oncotarget 2016; 7:56864-56875. [PMID: 27486754 PMCID: PMC5302958 DOI: 10.18632/oncotarget.10870] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Accepted: 07/10/2016] [Indexed: 12/14/2022] Open
Abstract
Cyclin-dependent kinases (CDKs) are potential cancer therapeutic targets because of their critical role in promoting cell growth. Dinaciclib is a novel CDK inhibitor currently under clinical evaluation for the treatment of advanced malignancies. In this study, we demonstrated the anti-tumor activity of dinaciclib in triple negative breast cancer (TNBC) patient derived xenograft (PDX) and cell lines in vitro and in vivo. Treatment with dinaciclib induced cell cycle arrest at G2/M phase and marked apoptosis. These changes were accompanied by reduced phosphorylation of CDK1 and retinoblastoma (Rb) protein and decreased protein levels of cyclin B1, cMYC and survivin. We further demonstrated that siRNA knockdown of CDK9, the kinase subunit of positive transcription elongation factor b (P-TEFb), instead of CDK1 or CDK2, reduced the levels of cyclin B1 and MYC in TNBC cell lines. These data support the importance of CDK9, in addition to CDK1, in mediating the growth inhibitory effect of dinaciclib in TNBC. Further investigation of CDK9 as a therapeutic target in TNBC is needed.
Collapse
Affiliation(s)
- Sandeep Rajput
- Section of Medical Oncology, Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Nimmish Khera
- Section of Medical Oncology, Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Zhanfang Guo
- Section of Medical Oncology, Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Jeremy Hoog
- Section of Medical Oncology, Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Shunqiang Li
- Section of Medical Oncology, Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, USA
- Siteman Cancer Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Cynthia X. Ma
- Section of Medical Oncology, Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, USA
- Siteman Cancer Center, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
46
|
Hu H, Wu J, Ao M, Wang H, Zhou T, Xue Y, Qiu Y, Fang M, Wu Z. Synthesis, structure-activity relationship studies and biological evaluation of novel 2,5-disubstituted indole derivatives as anticancer agents. Chem Biol Drug Des 2016; 88:766-778. [PMID: 27315790 DOI: 10.1111/cbdd.12808] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2016] [Revised: 06/05/2016] [Accepted: 06/12/2016] [Indexed: 01/13/2023]
Abstract
Three novel series of 2,5-disubstituted indole derivatives were synthesized and evaluated in vitro for their antiproliferative activity against human cancer cells and HIV-1 inhibition activity used as a readout of cellular activity. Most compounds were found to have potent anticancer activity. In particular, 2c and 3b which showed effectively to repress HIV-1 transcription had a pan antiproliferative activity in cervical cancer cells (HeLa), breast cancer cells (MCF-7), liver cancer cells (HepG2), and lung cancer cells (H460 and A549). While 3b exhibited high sensitivity to A549 cells with the IC50 value 0.48 ± 0.15 μm, 2c showed high selectivity toward HepG2 cells with the IC50 value 13.21 ± 0.30 μm. With respect to the cellular mechanism of action, HepG2 cells treated with 2c and A549 cells treated with 3b for 24 h were studied by annexin V/PI staining and Western blot analysis, and results revealed that 2c and 3b may induce cancer cells apoptosis through inhibiting the phosphorylation at Ser2 of RNAPII CTD which can be phosphorylated by cyclin-dependent kinase 9. These studies indicated that 2c and 3b may develop as potent lead compounds in the therapy of cancer. However, determining their roles in preventing HIV-1 still requires further intensive study.
Collapse
Affiliation(s)
- Hongyu Hu
- School of Pharmaceutical Sciences and the Key Laboratory for Chemical Biology of Fujian Province, Xiamen University, Xiamen, China
| | - Jun Wu
- School of Pharmaceutical Sciences and the Key Laboratory for Chemical Biology of Fujian Province, Xiamen University, Xiamen, China
| | - Mingtao Ao
- School of Pharmaceutical Sciences and the Key Laboratory for Chemical Biology of Fujian Province, Xiamen University, Xiamen, China
| | - Huiru Wang
- School of Pharmaceutical Sciences and the Key Laboratory for Chemical Biology of Fujian Province, Xiamen University, Xiamen, China
| | - Tongtong Zhou
- School of Pharmaceutical Sciences and the Key Laboratory for Chemical Biology of Fujian Province, Xiamen University, Xiamen, China
| | - Yuhua Xue
- School of Pharmaceutical Sciences and the Key Laboratory for Chemical Biology of Fujian Province, Xiamen University, Xiamen, China
| | - Yingkun Qiu
- School of Pharmaceutical Sciences and the Key Laboratory for Chemical Biology of Fujian Province, Xiamen University, Xiamen, China
| | - Meijuan Fang
- School of Pharmaceutical Sciences and the Key Laboratory for Chemical Biology of Fujian Province, Xiamen University, Xiamen, China.
| | - Zhen Wu
- School of Pharmaceutical Sciences and the Key Laboratory for Chemical Biology of Fujian Province, Xiamen University, Xiamen, China.
| |
Collapse
|
47
|
Cui J, Placzek WJ. PTBP1 modulation of MCL1 expression regulates cellular apoptosis induced by antitubulin chemotherapeutics. Cell Death Differ 2016; 23:1681-90. [PMID: 27367564 DOI: 10.1038/cdd.2016.60] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Revised: 04/30/2016] [Accepted: 05/30/2016] [Indexed: 01/01/2023] Open
Abstract
Myeloid cell leukemia sequence 1 (MCL1), an anti-apoptotic BCL2 family protein, is a key regulator of intrinsic apoptosis. Normal cells require strict control over MCL1 expression with aberrant MCL1 expression linked to the emergence of various diseases and chemoresistance. Previous studies have detailed how MCL1 expression is regulated by multiple mechanisms both transcriptionally and translationally. However, characterization of the post-transcriptional regulators of MCL1 mRNA is limited. Polypyrimidine tract binding protein 1 (PTBP1) is a known regulator of post-transcriptional gene expression that can control mRNA splicing, translation, stability and localization. Here we demonstrate that PTBP1 binds to MCL1 mRNA and that knockdown of PTBP1 upregulates MCL1 expression in cancer cells by stabilizing MCL1 mRNA and increasing MCL1 mRNA accumulation in cytoplasm. Further, we show that depletion of PTBP1 protects cancer cells from antitubulin agent-induced apoptosis in a MCL1-dependent manner. Taken together, our findings suggest that PTBP1 is a novel regulator of MCL1 mRNA by which it controls apoptotic response to antitubulin chemotherapeutics.
Collapse
Affiliation(s)
- J Cui
- Department of Biochemistry and Molecular Genetics, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - W J Placzek
- Department of Biochemistry and Molecular Genetics, The University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
48
|
Ayupe AC, Tahira AC, Camargo L, Beckedorff FC, Verjovski-Almeida S, Reis EM. Global analysis of biogenesis, stability and sub-cellular localization of lncRNAs mapping to intragenic regions of the human genome. RNA Biol 2016; 12:877-92. [PMID: 26151857 DOI: 10.1080/15476286.2015.1062960] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Long noncoding RNAs (lncRNAs) that map to intragenic regions of the human genome with the same (intronic lncRNAs) or opposite orientation (antisense lncRNAs) relative to protein-coding mRNAs have been largely dismissed from biochemical and functional characterization due to the belief that they are mRNA precursors, byproducts of RNA splicing or simply transcriptional noise. In this work, we used a custom microarray to investigate aspects of the biogenesis, processing, stability, evolutionary conservation, and cellular localization of ∼ 6,000 intronic lncRNAs and ∼ 10,000 antisense lncRNAs. Most intronic (2,903 of 3,427, 85%) and antisense lncRNAs (4,945 of 5,214, 95%) expressed in HeLa cells showed evidence of 5' cap modification, compatible with their transcription by RNAP II. Antisense lncRNAs (median t1/2 = 3.9 h) were significantly (p < 0.0001) more stable than mRNAs (median t1/2 = 3.2 h), whereas intronic lncRNAs (median t1/2 = 2.1 h) comprised a more heterogeneous class that included both stable (t1/2 > 3 h) and unstable (t1/2 < 1 h) transcripts. Intragenic lncRNAs display evidence of evolutionary conservation, have little/no coding potential and were ubiquitously detected in the cytoplasm. Notably, a fraction of the intronic and antisense lncRNAs (13 and 15%, respectively) were expressed from loci at which the corresponding host mRNA was not detected. The abundances of a subset of intronic/antisense lncRNAs were correlated (r ≥ |0.8|) with those of genes encoding proteins involved in cell division and DNA replication. Taken together, the findings of this study contribute novel biochemical and genomic information regarding intronic and antisense lncRNAs, supporting the notion that these classes include independently transcribed RNAs with potentials for exerting regulatory functions in the cell.
Collapse
Affiliation(s)
- Ana C Ayupe
- a Departamento de Bioquímica ; Instituto de Química ; Universidade de São Paulo ; Sao Paulo , Brazil
| | | | | | | | | | | |
Collapse
|
49
|
Finn RS, Aleshin A, Slamon DJ. Targeting the cyclin-dependent kinases (CDK) 4/6 in estrogen receptor-positive breast cancers. Breast Cancer Res 2016; 18:17. [PMID: 26857361 PMCID: PMC4746893 DOI: 10.1186/s13058-015-0661-5] [Citation(s) in RCA: 253] [Impact Index Per Article: 28.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2013] [Accepted: 12/10/2015] [Indexed: 12/18/2022] Open
Abstract
Despite significant advances in early detection and treatment, breast cancer still remains a major cause of morbidity and mortality for women. Our understanding of the molecular heterogeneity of the disease has significantly expanded over the past decade and the role of cell cycle signaling in both breast cancer oncogenesis and anti-estrogen resistance has gained increasing attention. The mammalian cell cycle is driven by a complex interplay between cyclins and their associated cyclin-dependent kinase (CDK) partners, and dysregulation of this process is one of the hallmarks of cancer. Despite this, initial results with broadly acting CDK inhibitors were largely disappointing. However, recent preclinical and phase I/II clinical studies using a novel, oral, reversible CDK4/6 inhibitor, palbociclib (PD-0332991), have validated the role of CDK4/6 as a potential target in estrogen receptor-positive (ER+) breast cancers. This review highlights our current understanding of CDK signaling in both normal and malignant breast tissues, with special attention placed on recent clinical advances in inhibition of CDK4/6 in ER+ disease.
Collapse
Affiliation(s)
- Richard S Finn
- Geffen School of Medicine at UCLA, Department of Medicine, Division of Hematology Oncology, 2825 Santa Monica Blvd, Santa Monica, CA, 90404, USA.
| | - Alexey Aleshin
- Geffen School of Medicine at UCLA, Department of Medicine, Division of Hematology Oncology, 2825 Santa Monica Blvd, Santa Monica, CA, 90404, USA
| | - Dennis J Slamon
- Stanford School of Medicine, 291 Campus Drive, Stanford, CA, 94305, USA
| |
Collapse
|
50
|
Enuka Y, Lauriola M, Feldman ME, Sas-Chen A, Ulitsky I, Yarden Y. Circular RNAs are long-lived and display only minimal early alterations in response to a growth factor. Nucleic Acids Res 2015; 44:1370-83. [PMID: 26657629 PMCID: PMC4756822 DOI: 10.1093/nar/gkv1367] [Citation(s) in RCA: 487] [Impact Index Per Article: 48.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Accepted: 11/24/2015] [Indexed: 12/20/2022] Open
Abstract
Circular RNAs (circRNAs) are widespread circles of non-coding RNAs with largely unknown function. Because stimulation of mammary cells with the epidermal growth factor (EGF) leads to dynamic changes in the abundance of coding and non-coding RNA molecules, and culminates in the acquisition of a robust migratory phenotype, this cellular model might disclose functions of circRNAs. Here we show that circRNAs of EGF-stimulated mammary cells are stably expressed, while mRNAs and microRNAs change within minutes. In general, the circRNAs we detected are relatively long-lived and weakly expressed. Interestingly, they are almost ubiquitously co-expressed with the corresponding linear transcripts, and the respective, shared promoter regions are more active compared to genes producing linear isoforms with no detectable circRNAs. These findings imply that altered abundance of circRNAs, unlike changes in the levels of other RNAs, might not play critical roles in signaling cascades and downstream transcriptional networks that rapidly commit cells to specific outcomes.
Collapse
Affiliation(s)
- Yehoshua Enuka
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Mattia Lauriola
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Morris E Feldman
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Aldema Sas-Chen
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Igor Ulitsky
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Yosef Yarden
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot 76100, Israel
| |
Collapse
|