1
|
Son YE, Cho HJ, Park HS. The MYB-like protein MylA contributes to conidiogenesis and conidial germination in Aspergillus nidulans. Commun Biol 2024; 7:768. [PMID: 38918572 PMCID: PMC11199622 DOI: 10.1038/s42003-024-05866-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 01/26/2024] [Indexed: 06/27/2024] Open
Abstract
Myeloblastosis (MYB)-like proteins are a family of highly conserved transcription factors in animals, plants, and fungi and are involved in the regulation of mRNA expression of genes. In this study, we identified and characterized one MYB-like protein in the model organism Aspergillus nidulans. We screened the mRNA levels of genes encoding MYB-like proteins containing two MYB repeats in conidia and found that the mRNA levels of four genes including flbD, cicD, and two uncharacterized genes, were high in conidia. To investigate the roles of two uncharacterized genes, AN4618 and AN10944, deletion mutants for each gene were generated. Our results revealed that AN4618 was required for fungal development. Therefore, we further investigated the role of AN4618, named as mylA, encoding the MYB-like protein containing two MYB repeats. Functional studies revealed that MylA was essential for normal fungal growth and development. Phenotypic and transcriptomic analyses demonstrated that deletion of mylA affected stress tolerance, cell wall integrity, and long-term viability in A. nidulans conidia. In addition, the germination rate of the mylA deletion mutant conidia was decreased compared with that of the wild-type conidia. Overall, this study suggests that MylA is critical for appropriate development, conidial maturation, dormancy, and germination in A. nidulans.
Collapse
Affiliation(s)
- Ye-Eun Son
- School of Food Science and Biotechnology, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - He-Jin Cho
- School of Food Science and Biotechnology, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Hee-Soo Park
- School of Food Science and Biotechnology, Kyungpook National University, Daegu, 41566, Republic of Korea.
- Department of Integrative Biology, Kyungpook National University, Daegu, 41566, Republic of Korea.
| |
Collapse
|
2
|
Hoch CC, Stögbauer F, Wollenberg B. Unraveling the Role of Epithelial-Mesenchymal Transition in Adenoid Cystic Carcinoma of the Salivary Glands: A Comprehensive Review. Cancers (Basel) 2023; 15:cancers15112886. [PMID: 37296849 DOI: 10.3390/cancers15112886] [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: 04/15/2023] [Revised: 05/21/2023] [Accepted: 05/23/2023] [Indexed: 06/12/2023] Open
Abstract
Salivary adenoid cystic carcinoma (SACC) is considered a challenging malignancy; it is characterized by a slow-growing nature, yet a high risk of recurrence and distant metastasis, presenting significant hurdles in its treatment and management. At present, there are no approved targeted agents available for the management of SACC and systemic chemotherapy protocols that have demonstrated efficacy remain to be elucidated. Epithelial-mesenchymal transition (EMT) is a complex process that is closely associated with tumor progression and metastasis, enabling epithelial cells to acquire mesenchymal properties, including increased mobility and invasiveness. Several molecular signaling pathways have been implicated in the regulation of EMT in SACC, and understanding these mechanisms is crucial to identifying new therapeutic targets and developing more effective treatment approaches. This manuscript aims to provide a comprehensive overview of the latest research on the role of EMT in SACC, including the molecular pathways and biomarkers involved in EMT regulation. By highlighting the most recent findings, this review offers insights into potential new therapeutic strategies that could improve the management of SACC patients, especially those with recurrent or metastatic disease.
Collapse
Affiliation(s)
- Cosima C Hoch
- Department of Otolaryngology, Head and Neck Surgery, School of Medicine, Technical University of Munich (TUM), 81675 Munich, Germany
| | - Fabian Stögbauer
- Institute of Pathology, School of Medicine, Technical University of Munich (TUM), 81675 Munich, Germany
| | - Barbara Wollenberg
- Department of Otolaryngology, Head and Neck Surgery, School of Medicine, Technical University of Munich (TUM), 81675 Munich, Germany
| |
Collapse
|
3
|
Odame E, Li L, Nabilla JA, Cai H, Xiao M, Ye J, Chen Y, Kyei B, Dai D, Zhan S, Cao J, Guo J, Zhong T, Wang L, Zhang H. miR-145-3p Inhibits MuSCs Proliferation and Mitochondria Mass via Targeting MYBL1 in Jianzhou Big-Eared Goats. Int J Mol Sci 2023; 24:ijms24098341. [PMID: 37176056 PMCID: PMC10179409 DOI: 10.3390/ijms24098341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 04/30/2023] [Accepted: 05/03/2023] [Indexed: 05/15/2023] Open
Abstract
Muscle growth and injury-induced regeneration are controlled by skeletal muscle satellite cells (MuSCs) through myogenesis in postnatal animals. Meanwhile, myogenesis is accompanied by mitochondrial function and enzyme activity. Nevertheless, the underlying molecular mechanisms involving non-coding RNAs including circular RNAs (circRNAs) and microRNAs (miRNAs) remain largely unsolved. Here, we explored the myogenic roles of miR-145-3p and MYBL1 on muscle development and mitochondrial mass. We noticed that overexpression of miR-145-3p inhibited MuSCs proliferation and reduced the number of viable cells. Meanwhile, deficiency of miR-145-3p caused by LNAantimiR-145-3p or an inhibitor retarded the differentiation of MuSCs. miR-145-3p altered the mitochondrial mass in MuSCs. Moreover, miR-145-3p targeted and negatively regulated the expression of CDR1as and MYBL1. The knockdown of the MYBL1 using ASO-2'MOE modification simulated the inhibitory function of miR-145-3p on cell proliferation. Additionally, MYBL1 mediated the regulation of miR-145-3p on Vexin, VCPIP1, COX1, COX2, and Pax7. These imply that CDR1as/miR-145-3p/MYBL1/COX1, COX2, VCPIP1/Vexin expression at least partly results in a reduction in mitochondrial mass and MuSCs proliferation. These novel findings confirm the importance of mitochondrial mass during myogenesis and the boosting of muscle/meat development in mammals.
Collapse
Affiliation(s)
- Emmanuel Odame
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - Li Li
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - Joshua Abdulai Nabilla
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - He Cai
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - Miao Xiao
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - Jiangfeng Ye
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - Yuan Chen
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - Bismark Kyei
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - Dinghui Dai
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - Siyuan Zhan
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - Jiaxue Cao
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - Jiazhong Guo
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - Tao Zhong
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - Linjie Wang
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - Hongping Zhang
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
| |
Collapse
|
4
|
Harada T, Perez MW, Kalfon J, Braes FD, Batley R, Eagle K, Nabet B, Leifer B, Kruell J, Paralkar VR, Stegmaier K, Koehler AN, Orkin SH, Pimkin M. Rapid-kinetics degron benchmarking reveals off-target activities and mixed agonism-antagonism of MYB inhibitors. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.07.536032. [PMID: 37066194 PMCID: PMC10104119 DOI: 10.1101/2023.04.07.536032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/18/2023]
Abstract
Attenuating aberrant transcriptional circuits holds great promise for the treatment of numerous diseases, including cancer. However, development of transcriptional inhibitors is hampered by the lack of a generally accepted functional cellular readout to characterize their target specificity and on-target activity. We benchmarked the direct gene-regulatory signatures of six agents reported as inhibitors of the oncogenic transcription factor MYB against targeted MYB degradation in a nascent transcriptomics assay. The inhibitors demonstrated partial specificity for MYB target genes but displayed significant off-target activity. Unexpectedly, the inhibitors displayed bimodal on-target effects, acting as mixed agonists-antagonists. Our data uncover unforeseen agonist effects of small molecules originally developed as TF inhibitors and argue that rapid-kinetics benchmarking against degron models should be used for functional characterization of transcriptional modulators.
Collapse
Affiliation(s)
- Taku Harada
- Cancer and Blood Disorders Center, Dana-Farber Cancer Institute and Boston Children’s Hospital, Harvard Medical School, Boston, MA, 02215, USA
| | - Monika W. Perez
- Cancer and Blood Disorders Center, Dana-Farber Cancer Institute and Boston Children’s Hospital, Harvard Medical School, Boston, MA, 02215, USA
| | - Jérémie Kalfon
- Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, MA, 02142, USA
| | - Flora Dievenich Braes
- Cancer and Blood Disorders Center, Dana-Farber Cancer Institute and Boston Children’s Hospital, Harvard Medical School, Boston, MA, 02215, USA
| | - Rashad Batley
- Cancer and Blood Disorders Center, Dana-Farber Cancer Institute and Boston Children’s Hospital, Harvard Medical School, Boston, MA, 02215, USA
| | - Kenneth Eagle
- Cancer and Blood Disorders Center, Dana-Farber Cancer Institute and Boston Children’s Hospital, Harvard Medical School, Boston, MA, 02215, USA
- Ken Eagle Consulting, Houston, TX, 77494, USA
| | - Behnam Nabet
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
| | - Becky Leifer
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| | - Jasmin Kruell
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| | - Vikram R. Paralkar
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Kimberly Stegmaier
- Cancer and Blood Disorders Center, Dana-Farber Cancer Institute and Boston Children’s Hospital, Harvard Medical School, Boston, MA, 02215, USA
- Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, MA, 02142, USA
| | - Angela N. Koehler
- Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, MA, 02142, USA
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Stuart H. Orkin
- Cancer and Blood Disorders Center, Dana-Farber Cancer Institute and Boston Children’s Hospital, Harvard Medical School, Boston, MA, 02215, USA
- Howard Hughes Medical Institute, Boston, MA, 02215, USA
| | - Maxim Pimkin
- Cancer and Blood Disorders Center, Dana-Farber Cancer Institute and Boston Children’s Hospital, Harvard Medical School, Boston, MA, 02215, USA
- Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, MA, 02142, USA
| |
Collapse
|
5
|
Tadi S, Ka-Yan Cheung V, Lee CS, Nguyen K, Luk PP, Low THH, Palme C, Clark J, Gupta R. MYB RNA detection by in situ hybridisation has high sensitivity and specificity for the diagnosis of adenoid cystic carcinoma. Pathology 2023; 55:456-465. [PMID: 37055331 DOI: 10.1016/j.pathol.2023.01.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Revised: 12/11/2022] [Accepted: 01/20/2023] [Indexed: 04/15/2023]
Abstract
Adenoid cystic carcinoma (ACC) is one of the most common primary salivary gland cancers. ACC has several benign and malignant mimics amongst salivary gland neoplasms. An accurate diagnosis of ACC is essential for optimal management of the patients and their follow-up. Upregulation of MYB has been described in 85-90% of ACC, but not in other salivary gland neoplasms. In ACC, MYB upregulation can occur as a result of a genetic rearrangement t(6;9) (q22-23;p23-24), MYB copy number variation (CNV), or enhancer hijacking of MYB. All mechanisms of MYB upregulation result in increased RNA transcription that can be detected using RNA in situ hybridisation (ISH) methods. In this study, utilising 138 primary salivary gland neoplasms including 78 ACC, we evaluate the diagnostic utility of MYB RNA ISH for distinguishing ACC from other primary salivary gland neoplasms with a prominent cribriform architecture including pleomorphic adenoma, basal cell adenoma, basal cell adenocarcinoma, epithelial myoepithelial carcinoma, and polymorphous adenocarcinoma. Fluorescent in situ hybridisation and next generation sequencing were also performed to evaluate the sensitivity and specificity of RNA ISH for detecting increased MYB RNA when MYB gene alterations were present. Detection of MYB RNA has 92.3% sensitivity and 98.2% specificity for a diagnosis of ACC amongst salivary gland neoplasms. The sensitivity of MYB RNA detection by ISH (92.3%) is significantly higher than that of the FISH MYB break-apart probe (42%) for ACC. Next generation sequencing did not demonstrate MYB alterations in cases that lacked MYB RNA overexpression indicating high sensitivity of MYB RNA ISH for detecting MYB gene alterations. The possibility that the sensitivity may be higher in clinical practice with contemporary samples as compared with older retrospective tissue samples with RNA degradation is not entirely excluded. In addition to the high sensitivity and specificity, MYB RNA testing can be performed using standard IHC platforms and protocols and evaluated using brightfield microscopy making it a time and cost-efficient diagnostic tool in routine clinical practice.
Collapse
Affiliation(s)
- Sahithi Tadi
- Department of Head and Neck Surgery, Chris O'Brien Lifehouse, Sydney, NSW, Australia
| | - Veronica Ka-Yan Cheung
- Department of Tissue Pathology and Diagnostic Oncology, NSW Health Pathology, Royal Prince Alfred Hospital, Sydney, NSW, Australia; Sydney Medical School, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - C Soon Lee
- Department of Tissue Pathology and Diagnostic Oncology, NSW Health Pathology, Royal Prince Alfred Hospital, Sydney, NSW, Australia; Sydney Medical School, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia; Discipline of Pathology, School of Medicine, Western Sydney University, Campbelltown, NSW, Australia; Department of Anatomical Pathology, Liverpool Hospital, Liverpool, NSW, Australia; Cancer Pathology Laboratory, Ingham Institute for Applied Medical Research, Liverpool, NSW, Australia; CONCERT Biobank, Ingham Institute for Applied Medical Research, Liverpool, NSW, Australia; South Western Sydney Clinical School, University of New South Wales, Liverpool, NSW, Australia
| | - Kevin Nguyen
- Department of Head and Neck Surgery, Chris O'Brien Lifehouse, Sydney, NSW, Australia; Department of Otolaryngology - Head and Neck Surgery, Faculty of Medicine and Health Sciences, Macquarie University, Macquarie Park, NSW, Australia
| | - Peter P Luk
- Department of Tissue Pathology and Diagnostic Oncology, NSW Health Pathology, Royal Prince Alfred Hospital, Sydney, NSW, Australia; Sydney Medical School, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Tsu-Hui Hubert Low
- Department of Head and Neck Surgery, Chris O'Brien Lifehouse, Sydney, NSW, Australia; Sydney Medical School, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia; Department of Otolaryngology - Head and Neck Surgery, Faculty of Medicine and Health Sciences, Macquarie University, Macquarie Park, NSW, Australia
| | - Carsten Palme
- Department of Head and Neck Surgery, Chris O'Brien Lifehouse, Sydney, NSW, Australia; Sydney Medical School, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Jonathan Clark
- Department of Head and Neck Surgery, Chris O'Brien Lifehouse, Sydney, NSW, Australia; Sydney Medical School, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia; Royal Prince Alfred Institute of Academic Surgery, Sydney Local Health District, Sydney, NSW, Australia
| | - Ruta Gupta
- Department of Tissue Pathology and Diagnostic Oncology, NSW Health Pathology, Royal Prince Alfred Hospital, Sydney, NSW, Australia; Sydney Medical School, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia.
| |
Collapse
|
6
|
Brayer KJ, Kang H, El-Naggar AK, Andreasen S, Homøe P, Kiss K, Mikkelsen L, Heegaard S, Pelaez D, Moeyersoms A, Tse DT, Guo Y, Lee DY, Ness SA. Dominant Gene Expression Profiles Define Adenoid Cystic Carcinoma (ACC) from Different Tissues: Validation of a Gene Signature Classifier for Poor Survival in Salivary Gland ACC. Cancers (Basel) 2023; 15:1390. [PMID: 36900183 PMCID: PMC10000625 DOI: 10.3390/cancers15051390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 02/11/2023] [Accepted: 02/17/2023] [Indexed: 02/25/2023] Open
Abstract
Adenoid cystic carcinoma (ACC) is an aggressive malignancy that most often arises in salivary or lacrimal glands but can also occur in other tissues. We used optimized RNA-sequencing to analyze the transcriptomes of 113 ACC tumor samples from salivary gland, lacrimal gland, breast or skin. ACC tumors from different organs displayed remarkedly similar transcription profiles, and most harbored translocations in the MYB or MYBL1 genes, which encode oncogenic transcription factors that may induce dramatic genetic and epigenetic changes leading to a dominant 'ACC phenotype'. Further analysis of the 56 salivary gland ACC tumors led to the identification of three distinct groups of patients, based on gene expression profiles, including one group with worse survival. We tested whether this new cohort could be used to validate a biomarker developed previously with a different set of 68 ACC tumor samples. Indeed, a 49-gene classifier developed with the earlier cohort correctly identified 98% of the poor survival patients from the new set, and a 14-gene classifier was almost as accurate. These validated biomarkers form a platform to identify and stratify high-risk ACC patients into clinical trials of targeted therapies for sustained clinical response.
Collapse
Affiliation(s)
- Kathryn J. Brayer
- Department of Internal Medicine, Division of Molecular Medicine, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA
- University of New Mexico Comprehensive Cancer Center, Albuquerque, NM 87131, USA
| | - Huining Kang
- University of New Mexico Comprehensive Cancer Center, Albuquerque, NM 87131, USA
- Department of Internal Medicine, Division of Epidemiology, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA
| | - Adel K. El-Naggar
- Department of Pathology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Simon Andreasen
- Department of Otorhinolaryngology and Maxillofacial Surgery, Zealand University Hospital, 4600 Køge, Denmark
| | - Preben Homøe
- Department of Otorhinolaryngology and Maxillofacial Surgery, Zealand University Hospital, 4600 Køge, Denmark
| | - Katalin Kiss
- Department of Pathology, Rigshospitalet, University of Copenhagen, 1165 Copenhagen, Denmark
| | - Lauge Mikkelsen
- Department of Pathology, Rigshospitalet, University of Copenhagen, 1165 Copenhagen, Denmark
- Department of Ophthalmology, Rigshospitalet-Glostrup, University of Copenhagen, 1165 Copenhagen, Denmark
| | - Steffen Heegaard
- Department of Ophthalmology, Rigshospitalet-Glostrup, University of Copenhagen, 1165 Copenhagen, Denmark
| | - Daniel Pelaez
- Dr. Nasser Al-Rashid Orbital Vision Research Center, Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Acadia Moeyersoms
- Dr. Nasser Al-Rashid Orbital Vision Research Center, Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- The Sheila and David Fuente Graduate Program in Cancer Biology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - David T. Tse
- Dr. Nasser Al-Rashid Orbital Vision Research Center, Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Yan Guo
- Department of Internal Medicine, Division of Molecular Medicine, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA
- University of New Mexico Comprehensive Cancer Center, Albuquerque, NM 87131, USA
| | - David Y. Lee
- University of New Mexico Comprehensive Cancer Center, Albuquerque, NM 87131, USA
- Department of Internal Medicine, Division of Hematology/Oncology, Section of Radiation Oncology, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA
| | - Scott A. Ness
- Department of Internal Medicine, Division of Molecular Medicine, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA
- University of New Mexico Comprehensive Cancer Center, Albuquerque, NM 87131, USA
| |
Collapse
|
7
|
Thakur S, Vasudev PG. MYB transcription factors and their role in Medicinal plants. Mol Biol Rep 2022; 49:10995-11008. [PMID: 36074230 DOI: 10.1007/s11033-022-07825-z] [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: 04/05/2022] [Revised: 07/06/2022] [Accepted: 07/27/2022] [Indexed: 11/29/2022]
Abstract
Transcription factors are multi-domain proteins that regulate gene expression in eukaryotic organisms. They are one of the largest families of proteins, which are structurally and functionally diverse. While there are transcription factors that are plant-specific, such as AP2/ERF, B3, NAC, SBP and WRKY, some transcription factors are present in both plants as well as other eukaryotic organisms. MYB transcription factors are widely distributed among all eukaryotes. In plants, the MYB transcription factors are involved in the regulation of numerous functions such as gene regulation in different metabolic pathways especially secondary metabolic pathways, regulation of different signalling pathways of plant hormones, regulation of genes involved in various developmental and morphological processes etc. Out of the thousands of MYB TFs that have been studied in plants, the majority of them have been studied in the model plants like Arabidopsis thaliana, Oryza sativa etc. The study of MYBs in other plants, especially medicinal plants, has been comparatively limited. But the increasing demand for medicinal plants for the production of biopharmaceuticals and important bioactive compounds has also increased the need to explore more number of these multifaceted transcription factors which play a significant role in the regulation of secondary metabolic pathways. These studies will ultimately contribute to medicinal plants' research and increased production of secondary metabolites, either through transgenic plants or through synthetic biology approaches. This review compiles studies on MYB transcription factors that are involved in the regulation of diverse functions in medicinal plants.
Collapse
Affiliation(s)
- Sudipa Thakur
- Plant Biotechnology Department, CSIR-Central Institute of Medicinal and Aromatic Plants, 226015, Lucknow, India.
| | - Prema G Vasudev
- Plant Biotechnology Department, CSIR-Central Institute of Medicinal and Aromatic Plants, 226015, Lucknow, India
| |
Collapse
|
8
|
Srivastava SK, Khan MA, Anand S, Zubair H, Deshmukh SK, Patel GK, Singh S, Andrews J, Wang B, Carter JE, Singh AP. MYB interacts with androgen receptor, sustains its ligand-independent activation and promotes castration resistance in prostate cancer. Br J Cancer 2022; 126:1205-1214. [PMID: 34837075 PMCID: PMC9023474 DOI: 10.1038/s41416-021-01641-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 10/27/2021] [Accepted: 11/10/2021] [Indexed: 11/08/2022] Open
Abstract
BACKGROUND Aberrant activation of androgen receptor signalling following castration therapy is a common clinical observation in prostate cancer (PCa). Earlier, we demonstrated the role of MYB overexpression in androgen-depletion resistance and PCa aggressiveness. Here, we investigated MYB-androgen receptor (AR) crosstalk and its functional significance. METHODS Interaction and co-localization of MYB and AR were examined by co-immunoprecipitation and immunofluorescence analyses, respectively. Protein levels were measured by immunoblot analysis and enzyme-linked immunosorbent assay. The role of MYB in ligand-independent AR transcriptional activity and combinatorial gene regulation was studied by promoter-reporter and chromatin immunoprecipitation assays. The functional significance of MYB in castration resistance was determined using an orthotopic mouse model. RESULTS MYB and AR interact and co-localize in the PCa cells. MYB-overexpressing PCa cells retain AR in the nucleus even when cultured under androgen-deprived conditions. AR transcriptional activity is also sustained in MYB-overexpressing cells in the absence of androgens. MYB binds and promotes AR occupancy to the KLK3 promoter. MYB-overexpressing PCa cells exhibit greater tumorigenicity when implanted orthotopically and quickly regain growth following castration leading to shorter mice survival, compared to those carrying low-MYB-expressing prostate tumours. CONCLUSIONS Our findings reveal a novel MYB-AR crosstalk in PCa and establish its role in castration resistance.
Collapse
Affiliation(s)
- Sanjeev Kumar Srivastava
- Department of Pathology, College of Medicine, University of South Alabama, Mobile, AL, 36617, USA
- Cancer Biology Program, Mitchell Cancer Institute, University of South Alabama, Mobile, AL, 36604, USA
| | - Mohammad Aslam Khan
- Department of Pathology, College of Medicine, University of South Alabama, Mobile, AL, 36617, USA
- Cancer Biology Program, Mitchell Cancer Institute, University of South Alabama, Mobile, AL, 36604, USA
| | - Shashi Anand
- Department of Pathology, College of Medicine, University of South Alabama, Mobile, AL, 36617, USA
- Cancer Biology Program, Mitchell Cancer Institute, University of South Alabama, Mobile, AL, 36604, USA
| | - Haseeb Zubair
- Department of Pathology, College of Medicine, University of South Alabama, Mobile, AL, 36617, USA
- Cancer Biology Program, Mitchell Cancer Institute, University of South Alabama, Mobile, AL, 36604, USA
| | - Sachin Kumar Deshmukh
- Department of Pathology, College of Medicine, University of South Alabama, Mobile, AL, 36617, USA
- Cancer Biology Program, Mitchell Cancer Institute, University of South Alabama, Mobile, AL, 36604, USA
| | - Girijesh Kumar Patel
- Department of Pathology, College of Medicine, University of South Alabama, Mobile, AL, 36617, USA
- Cancer Biology Program, Mitchell Cancer Institute, University of South Alabama, Mobile, AL, 36604, USA
| | - Seema Singh
- Department of Pathology, College of Medicine, University of South Alabama, Mobile, AL, 36617, USA
- Cancer Biology Program, Mitchell Cancer Institute, University of South Alabama, Mobile, AL, 36604, USA
- Department of Biochemistry and Molecular Biology, College of Medicine, University of South Alabama, Mobile, AL, 36688, USA
| | - Joel Andrews
- Bioimaging Core Facility, Mitchell Cancer Institute, University of South Alabama, Mobile, AL, 36604, USA
| | - Bin Wang
- Department of Mathematics and Statistics, University of South Alabama, Mobile, AL, 36688, USA
| | - James Elliot Carter
- Department of Pathology, College of Medicine, University of South Alabama, Mobile, AL, 36617, USA
| | - Ajay Pratap Singh
- Department of Pathology, College of Medicine, University of South Alabama, Mobile, AL, 36617, USA.
- Cancer Biology Program, Mitchell Cancer Institute, University of South Alabama, Mobile, AL, 36604, USA.
- Department of Biochemistry and Molecular Biology, College of Medicine, University of South Alabama, Mobile, AL, 36688, USA.
| |
Collapse
|
9
|
Transcription factor c-Myb: novel prognostic factor in osteosarcoma. Clin Exp Metastasis 2022; 39:375-390. [PMID: 34994868 DOI: 10.1007/s10585-021-10145-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Accepted: 12/28/2021] [Indexed: 12/21/2022]
Abstract
The transcription factor c-Myb is an oncoprotein promoting cell proliferation and survival when aberrantly activated/expressed, thus contributing to malignant transformation. Overexpression of c-Myb has been found in leukemias, breast, colon and adenoid cystic carcinoma. Recent studies revealed its expression also in osteosarcoma cell lines and suggested its functional importance during bone development. However, the relevance of c-Myb in control of osteosarcoma progression remains unknown. A retrospective clinical study was carried out to assess a relationship between c-Myb expression in archival osteosarcoma tissues and prognosis in a cohort of high-grade osteosarcoma patients. In addition, MYB was depleted in metastatic osteosarcoma cell lines SAOS-2 LM5 and 143B and their growth, chemosensitivity, migration and metastatic activity were determined. Immunohistochemical analysis revealed that high c-Myb expression was significantly associated with poor overall survival in the cohort and metastatic progression in young patients. Increased level of c-Myb was detected in metastatic osteosarcoma cell lines and its depletion suppressed their growth, colony-forming capacity, migration and chemoresistance in vitro in a cell line-dependent manner. MYB knock-out resulted in reduced metastatic activity of both SAOS-2 LM5 and 143B cell lines in immunodeficient mice. Transcriptomic analysis revealed the c-Myb-driven functional programs enriched for genes involved in the regulation of cell growth, stress response, cell adhesion and cell differentiation/morphogenesis. Wnt signaling pathway was identified as c-Myb target in osteosarcoma cells. Taken together, we identified c-Myb as a negative prognostic factor in osteosarcoma and showed its involvement in the regulation of osteosarcoma cell growth, chemosensitivity, migration and metastatic activity.
Collapse
|
10
|
Hu D, Shao W, Liu L, Wang Y, Yuan S, Liu Z, Liu J, Zhang J. Intricate crosstalk between MYB and noncoding RNAs in cancer. Cancer Cell Int 2021; 21:653. [PMID: 34876130 PMCID: PMC8650324 DOI: 10.1186/s12935-021-02362-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 11/24/2021] [Indexed: 11/10/2022] Open
Abstract
MYB is often overexpressed in malignant tumors and plays a carcinogenic role in the initiation and development of cancer. Deletion of the MYB regulatory C-terminal domain may be a driving mutation leading to tumorigenesis, therefore, different tumor mechanisms produce similar MYB proteins. As MYB is a transcription factor, priority has been given to identifying the genes that it regulates. All previous attention has been focused on protein-coding genes. However, an increasing number of studies have suggested that MYB can affect the complexity of cancer progression by regulating tumor-associated noncoding RNAs (ncRNAs), such as microRNAs, long-non-coding RNAs and circular RNAs. ncRNAs can regulate the expression of numerous downstream genes at the transcription, RNA processing and translation levels, thereby having various biological functions. Additionally, ncRNAs play important roles in regulating MYB expression. This review focuses on the intricate crosstalk between oncogenic MYB and ncRNAs, which play a pivotal role in tumorigenesis, including proliferation, apoptosis, angiogenesis, metastasis, senescence and drug resistance. In addition, we discuss therapeutic strategies for crosstalk between MYB and ncRNAs to prevent the occurrence and development of cancer.
Collapse
Affiliation(s)
- Dingyu Hu
- The First Affiliated Hospital, Department of Rheumatology, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Wenjun Shao
- The First Affiliated Hospital, Department of Rheumatology, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Li Liu
- The First Affiliated Hospital, Department of Rheumatology, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Yanyan Wang
- The First Affiliated Hospital, Department of Rheumatology, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Shunling Yuan
- The First Affiliated Hospital, Department of Rheumatology, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Zhaoping Liu
- The First Affiliated Hospital, Department of Rheumatology, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Jing Liu
- Hunan Province Key Laboratory of Basic and Applied Hematology, Molecular Biology Research Center & Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, 410078, Hunan, China.
| | - Ji Zhang
- The First Affiliated Hospital, Department of Rheumatology, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China. .,Department of Clinical Laboratory, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, 518033, Guangdong, China.
| |
Collapse
|
11
|
Lauxmann MA, Vazquez DS, Schilbert HM, Neubauer PR, Lammers KM, Dodero VI. From celiac disease to coccidia infection and vice-versa: The polyQ peptide CXCR3-interaction axis. Bioessays 2021; 43:e2100101. [PMID: 34705290 DOI: 10.1002/bies.202100101] [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: 04/13/2021] [Revised: 09/04/2021] [Accepted: 10/06/2021] [Indexed: 11/11/2022]
Abstract
Zonulin is a physiological modulator of intercellular tight junctions, which upregulation is involved in several diseases like celiac disease (CeD). The polyQ gliadin fragment binds to the CXCR3 chemokine receptor that activates zonulin upregulation, leading to increased intestinal permeability in humans. Here, we report a general hypothesis based on the structural connection between the polyQ sequence of the immunogenic CeD protein, gliadin, and enteric coccidian parasites proteins. Firstly, a novel interaction pathway between the parasites and the host is described based on the structural similarities between polyQ gliadin fragments and the parasite proteins. Secondly, a potential connection between coccidial infections as a novel environmental trigger of CeD is hypothesized. Therefore, this report represents a promising breakthrough for coccidian research and points out the potential role of coccidian parasites as a novel trigger of CeD that might define a preventive strategy for gluten-related disorders in general. Also see the video abstract here: https://youtu.be/oMaQasStcFI.
Collapse
Affiliation(s)
- Martin A Lauxmann
- Institute for Biochemistry, Brandenburg Medical School (MHB) Theodor Fontane, Germany.,Department of Nephrology, Campus Clinic Brandenburg, Brandenburg Medical School (MHB) Theodor Fontane, Germany
| | - Diego S Vazquez
- Grupo de Biología Estructural y Biotecnología (GBEyB-IMBICE), Departamento de Ciencia y Tecnología, Universidad Nacional de Quilmes, Bernal, Buenos Aires, Argentina.,Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Ciudad Autónoma de Buenos Aires, Argentina
| | - Hanna M Schilbert
- Department of Chemistry, Organic Chemistry OCIII, Universität Bielefeld, Universitätsstraße 25, Bielefeld, Germany.,Genetics and Genomics of Plants, Center for Biotechnology (CeBiTec) & Faculty of Biology, Universitätsstraße 25, Bielefeld, 33615, Germany
| | - Pia R Neubauer
- Department of Chemistry, Organic Chemistry OCIII, Universität Bielefeld, Universitätsstraße 25, Bielefeld, Germany
| | | | - Veronica I Dodero
- Department of Chemistry, Organic Chemistry OCIII, Universität Bielefeld, Universitätsstraße 25, Bielefeld, Germany
| |
Collapse
|
12
|
Śledzińska P, Bebyn MG, Furtak J, Kowalewski J, Lewandowska MA. Prognostic and Predictive Biomarkers in Gliomas. Int J Mol Sci 2021; 22:ijms221910373. [PMID: 34638714 PMCID: PMC8508830 DOI: 10.3390/ijms221910373] [Citation(s) in RCA: 131] [Impact Index Per Article: 43.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 09/14/2021] [Accepted: 09/22/2021] [Indexed: 12/17/2022] Open
Abstract
Gliomas are the most common central nervous system tumors. New technologies, including genetic research and advanced statistical methods, revolutionize the therapeutic approach to the patient and reveal new points of treatment options. Moreover, the 2021 World Health Organization Classification of Tumors of the Central Nervous System has fundamentally changed the classification of gliomas and incorporated many molecular biomarkers. Given the rapid progress in neuro-oncology, here we compile the latest research on prognostic and predictive biomarkers in gliomas. In adult patients, IDH mutations are positive prognostic markers and have the greatest prognostic significance. However, CDKN2A deletion, in IDH-mutant astrocytomas, is a marker of the highest malignancy grade. Moreover, the presence of TERT promoter mutations, EGFR alterations, or a combination of chromosome 7 gain and 10 loss upgrade IDH-wildtype astrocytoma to glioblastoma. In pediatric patients, H3F3A alterations are the most important markers which predict the worse outcome. MGMT promoter methylation has the greatest clinical significance in predicting responses to temozolomide (TMZ). Conversely, mismatch repair defects cause hypermutation phenotype predicting poor response to TMZ. Finally, we discussed liquid biopsies, which are promising diagnostic, prognostic, and predictive techniques, but further work is needed to implement these novel technologies in clinical practice.
Collapse
Affiliation(s)
- Paulina Śledzińska
- Department of Thoracic Surgery and Tumors, Ludwik Rydygier Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, 85-067 Torun, Poland
- The F. Lukaszczyk Oncology Center, Molecular Oncology and Genetics Department, Innovative Medical Forum, 85-796 Bydgoszcz, Poland
| | - Marek G Bebyn
- The F. Lukaszczyk Oncology Center, Molecular Oncology and Genetics Department, Innovative Medical Forum, 85-796 Bydgoszcz, Poland
- Faculty of Medicine, Medical University of Gdańsk, 80-210 Gdańsk, Poland
| | - Jacek Furtak
- Department of Neurosurgery, 10th Military Research Hospital and Polyclinic, 85-681 Bydgoszcz, Poland
- Franciszek Lukaszczyk Oncology Center, Department of Neurooncology and Radiosurgery, 85-796 Bydgoszcz, Poland
| | - Janusz Kowalewski
- Department of Thoracic Surgery and Tumors, Ludwik Rydygier Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, 85-067 Torun, Poland
| | - Marzena A Lewandowska
- Department of Thoracic Surgery and Tumors, Ludwik Rydygier Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, 85-067 Torun, Poland
- The F. Lukaszczyk Oncology Center, Molecular Oncology and Genetics Department, Innovative Medical Forum, 85-796 Bydgoszcz, Poland
| |
Collapse
|
13
|
Pediatric Glioma: An Update of Diagnosis, Biology, and Treatment. Cancers (Basel) 2021; 13:cancers13040758. [PMID: 33673070 PMCID: PMC7918156 DOI: 10.3390/cancers13040758] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2021] [Revised: 02/08/2021] [Accepted: 02/08/2021] [Indexed: 01/03/2023] Open
Abstract
Simple Summary Recent research has enhanced our understanding of the diverse biological processes that occur in pediatric gliomas; and molecular genetic analysis has become essential to diagnose and treat these conditions. Because targetable molecular aberrations can be detected in pediatric gliomas, identifying these aberrations is very important. This review provides an overview of pediatric gliomas, and describes recent developments made in strategies for their diagnosis and treatment. Additionally, it presents a current picture of pediatric gliomas in light of advances in molecular genetics, and describes the current scientific progress in gliomas’ treatment using information from recently completed and ongoing clinical trials. The era of incorporating molecular genetic analysis into clinical practice is emerging. Abstract Recent research has promoted elucidation of the diverse biological processes that occur in pediatric central nervous system (CNS) tumors. Molecular genetic analysis is essential not only for proper classification, but also for monitoring biological behavior and clinical management of tumors. Ever since the 2016 World Health Organization classification of CNS tumors, molecular profiling has become an indispensable step in the diagnosis, prediction of prognosis, and treatment of pediatric as well as adult CNS tumors. These molecular data are changing diagnosis, leading to new guidelines, and offering novel molecular targeted therapies. The Consortium to Inform Molecular and Practical Approaches to CNS Tumor Taxonomy (cIMPACT-NOW) makes practical recommendations using recent advances in CNS tumor classification, particularly in molecular discernment of these neoplasms as morphology-based classification of tumors is being replaced by molecular-based classification. In this article, we summarize recent knowledge to provide an overview of pediatric gliomas, which are major pediatric CNS tumors, and describe recent developments in strategies employed for their diagnosis and treatment.
Collapse
|
14
|
Caballero-Palacios MC, Villegas-Ruiz V, Ramírez-Chiquito JC, Medina-Vera I, Zapata-Tarres M, Mojica-Espinosa R, Cárdenas-Cardos R, Paredes-Aguilera R, Rivera-Luna R, Juárez-Méndez S. v-myb avian myeloblastosis viral oncogene homolog expression is a potential molecular diagnostic marker for B-cell acute lymphoblastic leukemia. Asia Pac J Clin Oncol 2020; 17:60-67. [PMID: 32779388 DOI: 10.1111/ajco.13406] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Accepted: 05/28/2020] [Indexed: 11/30/2022]
Abstract
BACKGROUND B-cell acute lymphoblastic leukemia (B-ALL) is the most commonly diagnosed childhood malignancy worldwide and is especially common in Mexico. Additionally, the number of cases has increased in recent years. Thus, it is very important to develop molecular strategies to diagnose leukemia. The aim of this study was to investigate MYB expression and to determine its impact on the diagnosis of B-ALL. METHODS We analyzed the B-ALL gene expression profile by microarray data mining. Bioinformatics analysis was performed to identify the genes that are overexpressed in leukemia. We determined that MYB was highly expressed in leukemia. Then, we validated MYB expression in 70 patients with B-ALL and in 16 healthy controls (HCs) using qRT-PCR. The results were statistically analyzed using the Kolmogorov-Smirnov Z test, Mann-Whitney U test, receiver operating characteristic curves, and the Youden index. RESULTS The microarrays showed that MYB was overexpressed in B-ALL patients with a fold change of 57.8728 and a P value of 2.56-195 . MYB expression showed great variability among the patients analyzed. However, compared to the HCs, the B-ALL patients had a P value < .0001, an area under the curve of 0.813, and a Youden index of 1.46, indicating the statistical significance. CONCLUSION MYB expression in B-ALL cells could be a potential molecular marker for childhood leukemia.
Collapse
Affiliation(s)
| | - Vanessa Villegas-Ruiz
- Experimental Oncology Laboratory, Research Department, National Institute of Pediatrics, Mexico City, Mexico
| | | | - Isabel Medina-Vera
- Research Methodology Department, National Institute of Pediatrics, Mexico City, Mexico
| | - Martha Zapata-Tarres
- Department of Pediatric Oncology, National Institute of Pediatrics, Mexico City, Mexico
| | | | - Rocio Cárdenas-Cardos
- Department of Pediatric Oncology, National Institute of Pediatrics, Mexico City, Mexico
| | | | - Roberto Rivera-Luna
- Division of Pediatric Hemato/Oncology, National Institute of Pediatrics, Mexico City, Mexico
| | - Sergio Juárez-Méndez
- Experimental Oncology Laboratory, Research Department, National Institute of Pediatrics, Mexico City, Mexico
| |
Collapse
|
15
|
Gažová I, Lefevre L, Bush SJ, Clohisey S, Arner E, de Hoon M, Severin J, van Duin L, Andersson R, Lengeling A, Hume DA, Summers KM. The Transcriptional Network That Controls Growth Arrest and Macrophage Differentiation in the Human Myeloid Leukemia Cell Line THP-1. Front Cell Dev Biol 2020; 8:498. [PMID: 32719792 PMCID: PMC7347797 DOI: 10.3389/fcell.2020.00498] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Accepted: 05/25/2020] [Indexed: 12/12/2022] Open
Abstract
The response of the human acute myeloid leukemia cell line THP-1 to phorbol esters has been widely studied to test candidate leukemia therapies and as a model of cell cycle arrest and monocyte-macrophage differentiation. Here we have employed Cap Analysis of Gene Expression (CAGE) to analyze a dense time course of transcriptional regulation in THP-1 cells treated with phorbol myristate acetate (PMA) over 96 h. PMA treatment greatly reduced the numbers of cells entering S phase and also blocked cells exiting G2/M. The PMA-treated cells became adherent and expression of mature macrophage-specific genes increased progressively over the duration of the time course. Within 1–2 h PMA induced known targets of tumor protein p53 (TP53), notably CDKN1A, followed by gradual down-regulation of cell-cycle associated genes. Also within the first 2 h, PMA induced immediate early genes including transcription factor genes encoding proteins implicated in macrophage differentiation (EGR2, JUN, MAFB) and down-regulated genes for transcription factors involved in immature myeloid cell proliferation (MYB, IRF8, GFI1). The dense time course revealed that the response to PMA was not linear and progressive. Rather, network-based clustering of the time course data highlighted a sequential cascade of transient up- and down-regulated expression of genes encoding feedback regulators, as well as transcription factors associated with macrophage differentiation and their inferred target genes. CAGE also identified known and candidate novel enhancers expressed in THP-1 cells and many novel inducible genes that currently lack functional annotation and/or had no previously known function in macrophages. The time course is available on the ZENBU platform allowing comparison to FANTOM4 and FANTOM5 data.
Collapse
Affiliation(s)
- Iveta Gažová
- The Roslin Institute, The University of Edinburgh, Edinburgh, United Kingdom
| | - Lucas Lefevre
- The Roslin Institute, The University of Edinburgh, Edinburgh, United Kingdom
| | - Stephen J Bush
- The Roslin Institute, The University of Edinburgh, Edinburgh, United Kingdom
| | - Sara Clohisey
- The Roslin Institute, The University of Edinburgh, Edinburgh, United Kingdom
| | - Erik Arner
- RIKEN Center for Integrative Medical Sciences, Kanagawa, Yokohama, Japan
| | - Michiel de Hoon
- RIKEN Center for Integrative Medical Sciences, Kanagawa, Yokohama, Japan
| | - Jessica Severin
- RIKEN Center for Integrative Medical Sciences, Kanagawa, Yokohama, Japan
| | - Lucas van Duin
- Bioinformatics Centre, University of Copenhagen, Copenhagen, Denmark
| | - Robin Andersson
- Bioinformatics Centre, University of Copenhagen, Copenhagen, Denmark
| | | | - David A Hume
- Mater Research Institute - University of Queensland, Translational Research Institute, Brisbane, QLD, Australia
| | - Kim M Summers
- The Roslin Institute, The University of Edinburgh, Edinburgh, United Kingdom.,Mater Research Institute - University of Queensland, Translational Research Institute, Brisbane, QLD, Australia
| |
Collapse
|
16
|
de Blank P, Fouladi M, Huse JT. Molecular markers and targeted therapy in pediatric low-grade glioma. J Neurooncol 2020; 150:5-15. [PMID: 32399739 DOI: 10.1007/s11060-020-03529-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Accepted: 05/04/2020] [Indexed: 12/14/2022]
Abstract
INTRODUCTION Recently discovered molecular alterations in pediatric low-grade glioma have helped to refine the classification of these tumors and offered novel targets for therapy. Genetic aberrations may combine with histopathology to offer new insights into glioma classification, gliomagenesis and prognosis. Therapies targeting common genetic aberrations in the MAPK pathway offer a novel mechanism of tumor control that is currently under study. METHODS We have reviewed common molecular alterations found in pediatric low-grade glioma as well as recent clinical trials of MEK and BRAF inhibitors. RESULTS In this topic review, we examine the current understanding of molecular alterations in pediatric low-grade glioma, as well as their role in diagnosis, prognosis and therapy. We summarize current data on the efficacy of targeted therapies in pediatric low-grade gliomas, as well as the many unanswered questions that these new discoveries and therapies raise. CONCLUSIONS The identification of driver alterations in pediatric low-grade glioma and the development of targeted therapies have opened new therapeutic avenues for patients with low-grade gliomas.
Collapse
Affiliation(s)
- Peter de Blank
- Department of Pediatrics, University of Cincinnati College of Medicine and the Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH, 45229, USA.
| | - Maryam Fouladi
- Department of Pediatrics, University of Cincinnati College of Medicine and the Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH, 45229, USA
| | - Jason T Huse
- Departments of Pathology and Translational Molecular Pathology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
17
|
Huang FL, Liao EC, Li CL, Yen CY, Yu SJ. Pathogenesis of pediatric B-cell acute lymphoblastic leukemia: Molecular pathways and disease treatments. Oncol Lett 2020; 20:448-454. [PMID: 32565969 PMCID: PMC7285861 DOI: 10.3892/ol.2020.11583] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2019] [Accepted: 04/03/2020] [Indexed: 01/12/2023] Open
Abstract
B-cell acute lymphoblastic lymphoma (B-ALL) is a disease found mainly in children and in young adults. B-ALL is characterized by the rapid proliferation of poorly differentiated lymphoid progenitor cells inside the bone marrow. In the United States, ~4,000 of these patients are diagnosed each year, accounting for ~30% of childhood cancer types. The tumorigenesis of the disease involves a number of abnormal gene expressions (including TEL-AML1, BCR-ABL-1, RAS and PI3K) leading to dysregulated cell cycle. Risk factors of B-ALL are the history of parvovirus B 19 infection, high birth weight and exposure to environmental toxins. These risk factors can induce abnormal DNA methylation and DNA damages. Treatment procedures are divided into three phases: Induction, consolidation and maintenance. The goal of treatment is complete remission without relapses. Apart from traditional treatments, newly developed approaches include gene targeting therapy, with the aim of wiping out leukemic cells through the inhibition of mitogen-activated protein kinases and via c-Myb inhibition enhancing sensitivity to chemotherapy. To evaluate the efficacy of ongoing treatments, several indicators are currently used. The indicators include the expression levels of microRNAs (miRs) miR-146a, miR-155, miR-181a and miR-195, and soluble interleukin 2 receptor. Multiple drug resistance and levels of glutathione reductase can affect treatment efficacy through the increased efflux of anti-cancer drugs and weakening the effect of chemotherapy through the reduction of intracellular reactive oxygen species. The present review appraised recent studies on B-ALL regarding its pathogenesis, risk factors, treatments, treatment evaluation and causes of disease relapse. Understanding the mechanisms of B-ALL initiation and causes of treatment failure can help physicians improve disease management and reduce relapses.
Collapse
Affiliation(s)
- Fang-Liang Huang
- Children's Medical Center, Taichung Veterans General Hospital, Xitun, Taichung 40705, Taiwan, R.O.C.,Department of Physical Therapy, Hungkuang University, Shalu, Taichung 433, Taiwan, R.O.C
| | - En-Chih Liao
- Department of Medicine, Mackay Medical College, Sanzhi, New Taipei 252, Taiwan, R.O.C
| | - Chia-Ling Li
- Children's Medical Center, Taichung Veterans General Hospital, Xitun, Taichung 40705, Taiwan, R.O.C
| | - Chung-Yang Yen
- Department of Dermatology, Taichung Veterans General Hospital, Xitun, Taichung 40705, Taiwan, R.O.C
| | - Sheng-Jie Yu
- Department of Medical Education and Research, Kaohsiung Veterans General Hospital, Zuoying, Kaohsiung 813, Taiwan, R.O.C
| |
Collapse
|
18
|
Ryall S, Tabori U, Hawkins C. Pediatric low-grade glioma in the era of molecular diagnostics. Acta Neuropathol Commun 2020; 8:30. [PMID: 32164789 PMCID: PMC7066826 DOI: 10.1186/s40478-020-00902-z] [Citation(s) in RCA: 187] [Impact Index Per Article: 46.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Accepted: 02/21/2020] [Indexed: 12/17/2022] Open
Abstract
Low grade gliomas are the most frequent brain tumors in children and encompass a spectrum of histologic entities which are currently assigned World Health Organisation grades I and II. They differ substantially from their adult counterparts in both their underlying genetic alterations and in the infrequency with which they transform to higher grade tumors. Nonetheless, children with low grade glioma are a therapeutic challenge due to the heterogeneity in their clinical behavior – in particular, those with incomplete surgical resection often suffer repeat progressions with resultant morbidity and, in some cases, mortality. The identification of up-regulation of the RAS–mitogen-activated protein kinase (RAS/MAPK) pathway as a near universal feature of these tumors has led to the development of targeted therapeutics aimed at improving responses while mitigating patient morbidity. Here, we review how molecular information can help to further define the entities which fall under the umbrella of pediatric-type low-grade glioma. In doing so we discuss the specific molecular drivers of pediatric low grade glioma and how to effectively test for them, review the newest therapeutic agents and their utility in treating this disease, and propose a risk-based stratification system that considers both clinical and molecular parameters to aid clinicians in making treatment decisions.
Collapse
|
19
|
The Tumor Suppressor Roles of MYBBP1A, a Major Contributor to Metabolism Plasticity and Stemness. Cancers (Basel) 2020; 12:cancers12010254. [PMID: 31968688 PMCID: PMC7017249 DOI: 10.3390/cancers12010254] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Revised: 12/18/2019] [Accepted: 01/14/2020] [Indexed: 12/12/2022] Open
Abstract
The MYB binding protein 1A (MYBBP1A, also known as p160) acts as a co-repressor of multiple transcription factors involved in many physiological processes. Therefore, MYBBP1A acts as a tumor suppressor in multiple aspects related to cell physiology, most of them very relevant for tumorigenesis. We explored the different roles of MYBBP1A in different aspects of cancer, such as mitosis, cellular senescence, epigenetic regulation, cell cycle, metabolism plasticity and stemness. We especially reviewed the relationships between MYBBP1A, the inhibitory role it plays by binding and inactivating c-MYB and its regulation of PGC-1α, leading to an increase in the stemness and the tumor stem cell population. In addition, MYBBP1A causes the activation of PGC-1α directly and indirectly through c-MYB, inducing the metabolic change from glycolysis to oxidative phosphorylation (OXPHOS). Therefore, the combination of these two effects caused by the decreased expression of MYBBP1A provides a selective advantage to tumor cells. Interestingly, this only occurs in cells lacking pVHL. Finally, the loss of MYBBP1A occurs in 8%–9% of renal tumors. tumors, and this subpopulation could be studied as a possible target of therapies using inhibitors of mitochondrial respiration.
Collapse
|
20
|
Gorbatenko A, Søkilde R, Sorensen EE, Newie I, Persson H, Morancho B, Arribas J, Litman T, Rovira C, Pedersen SF. HER2 and p95HER2 differentially regulate miRNA expression in MCF-7 breast cancer cells and downregulate MYB proteins through miR-221/222 and miR-503. Sci Rep 2019; 9:3352. [PMID: 30833639 PMCID: PMC6399295 DOI: 10.1038/s41598-019-39733-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Accepted: 02/04/2019] [Indexed: 12/18/2022] Open
Abstract
The HER2 oncogene and its truncated form p95HER2 play central roles in breast cancer. Here, we show that although HER2 and p95HER2 generally elicit qualitatively similar changes in miRNA profile in MCF-7 breast cancer cells, a subset of changes are distinct and p95HER2 shifts the miRNA profile towards the basal breast cancer subtype. High-throughput miRNA profiling was carried out 15, 36 and 60 h after HER2 or p95HER2 expression and central hits validated by RT-qPCR. miRNAs strongly regulated by p95HER2 yet not by HER2, included miR-221, miR-222, miR-503, miR-29a, miR-149, miR-196 and miR-361. Estrogen receptor-α (ESR1) expression was essentially ablated by p95HER2 expression, in a manner recapitulated by miR-221/-222 mimics. c-Myb family transcription factors MYB and MYBL1, but not MYBL2, were downregulated by p95HER2 and by miR-503 or miR-221/-222 mimics. MYBL1 3′UTR inhibition by miR-221/222 was lost by deletion of a single putative miR-221/222 binding sites. p95HER2 expression, or knockdown of either MYB protein, elicited upregulation of tissue inhibitor of matrix metalloprotease-2 (TIMP2). miR-221/222 and -503 mimics increased, and TIMP2 knockdown decreased, cell migration and invasion. A similar pathway was operational in T47D- and SKBr-3 cells. This work reveals important differences between HER2- and p95HER2- mediated miRNA changes in breast cancer cells, provides novel mechanistic insight into regulation of MYB family transcription factors by p95HER2, and points to a role for a miR-221/222– MYB family–TIMP2 axis in regulation of motility in breast cancer cells.
Collapse
Affiliation(s)
- Andrej Gorbatenko
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, New York, 10029, USA.,Section for Cell Biology and Physiology, Department of Biology, Faculty of Science, University of Copenhagen, Universitetsparken 13, DK-2100, Copenhagen, Denmark
| | - Rolf Søkilde
- BioCare, Strategic Cancer Research Program, Lund, Sweden.,Department of Clinical Sciences Lund, Oncology and Pathology, Faculty of Medicine, Lund University, Lund, Sweden
| | - Ester E Sorensen
- Section for Cell Biology and Physiology, Department of Biology, Faculty of Science, University of Copenhagen, Universitetsparken 13, DK-2100, Copenhagen, Denmark
| | - Inga Newie
- BioCare, Strategic Cancer Research Program, Lund, Sweden.,Department of Clinical Sciences Lund, Oncology and Pathology, Faculty of Medicine, Lund University, Lund, Sweden
| | - Helena Persson
- BioCare, Strategic Cancer Research Program, Lund, Sweden.,Department of Clinical Sciences Lund, Oncology and Pathology, Faculty of Medicine, Lund University, Lund, Sweden
| | - Beatriz Morancho
- Preclinical Research Program, Vall d'Hebron Institute of Oncology and CIBERONC, 08035, Barcelona, Spain
| | - Joaquin Arribas
- Preclinical Research Program, Vall d'Hebron Institute of Oncology and CIBERONC, 08035, Barcelona, Spain.,Department of Biochemistry and Molecular Biology, Universitat Autonoma de Barcelona, Campus de la UAB, JA, Bellaterra, Spain.,Institució Catalana de Recerca i Estudis Avançats, JA, Barcelona, Spain
| | - Thomas Litman
- Department of International Health, Immunology and Microbiology, University of Copenhagen, Blegdamsvej 3B, DK-2200, Copenhagen, Denmark
| | - Carlos Rovira
- BioCare, Strategic Cancer Research Program, Lund, Sweden.,Department of Clinical Sciences Lund, Oncology and Pathology, Faculty of Medicine, Lund University, Lund, Sweden
| | - Stine Falsig Pedersen
- Section for Cell Biology and Physiology, Department of Biology, Faculty of Science, University of Copenhagen, Universitetsparken 13, DK-2100, Copenhagen, Denmark.
| |
Collapse
|
21
|
Loss of MYBBP1A Induces Cancer Stem Cell Activity in Renal Cancer. Cancers (Basel) 2019; 11:cancers11020235. [PMID: 30781655 PMCID: PMC6406377 DOI: 10.3390/cancers11020235] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 02/03/2019] [Accepted: 02/13/2019] [Indexed: 01/20/2023] Open
Abstract
Tumors are cellular ecosystems where different populations and subpopulations of cells coexist. Among these cells, cancer stem cells (CSCs) are considered to be the origin of the tumor mass, being involved in metastasis and in the resistance to conventional therapies. Furthermore, tumor cells have an enormous plasticity and a phenomenon of de-differentiation of mature tumor cells to CSCs may occur. Therefore, it is essential to identify genetic alterations that cause the de-differentiation of mature tumor cells to CSCs for the future design of therapeutic strategies. In this study, we characterized the role of MYBBP1A by experiments in cell lines, xenografts and human tumor samples. We have found that MYBBP1A downregulation increases c-MYB (Avian myeloblastosis viral oncogene homolog) activity, leading to a rise in the stem-like cell population. We identified that the downregulation of MYBBP1A increases tumorigenic properties, in vitro and in vivo, in renal carcinoma cell lines that express high levels of c-MYB exclusively. Moreover, in a cohort of renal tumors, MYBBP1A is downregulated or lost in a significant percentage of tumors correlating with poor patient prognosis and a metastatic tendency. Our data support the role of MYBBP1A as a tumor suppressor by repressing c-MYB, acting as an important regulator of the plasticity of tumor cells.
Collapse
|
22
|
Abstract
The c-Myb gene encodes a transcription factor that regulates cell proliferation, differentiation, and apoptosis through protein-protein interaction and transcriptional regulation of signaling pathways. The protein is frequently overexpressed in human leukemias, breast cancers, and other solid tumors suggesting that it is a bona fide oncogene. c-MYB is often overexpressed by translocation in human tumors with t(6;7)(q23;q34) resulting in c-MYB-TCRβ in T cell ALL, t(X;6)(p11;q23) with c-MYB-GATA1 in acute basophilic leukemia, and t(6;9)(q22-23;p23-24) with c-MYB-NF1B in adenoid cystic carcinoma. Antisense oligonucleotides to c-MYB were developed to purge bone marrow cells to eliminate tumor cells in leukemias. Recently, small molecules that inhibit c-MYB activity have been developed to disrupt its interaction with p300. The Dmp1 (cyclin D binding myb-like protein 1; Dmtf1) gene was isolated through its virtue for binding to cyclin D2. It is a transcription factor that has a Myb-like repeat for DNA binding. The Dmtf1 protein directly binds to the Arf promoter for transactivation and physically interacts with p53 to activate the p53 pathway. The gene is hemizygously deleted in 35-42% of human cancers and is associated with longer survival. The significances of aberrant expression of c-MYB and DMTF1 proteins in human cancers and their clinical significances are discussed.
Collapse
Affiliation(s)
- Elizabeth A. Fry
- The Department of Pathology, Wake Forest University School of Medicine, Medical Center Blvd., Winston-Salem, NC 27157 USA
| | - Kazushi Inoue
- The Department of Pathology, Wake Forest University School of Medicine, Medical Center Blvd., Winston-Salem, NC 27157 USA
| |
Collapse
|
23
|
Association of SHMT1, MAZ, ERG, and L3MBTL3 Gene Polymorphisms with Susceptibility to Multiple Sclerosis. Biochem Genet 2018; 57:355-370. [PMID: 30456721 DOI: 10.1007/s10528-018-9894-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Accepted: 11/07/2018] [Indexed: 01/08/2023]
Abstract
Multiple sclerosis (MS) is the most common inflammatory and chronic disease of the central nervous system (CNS). A complex interaction between genetic, environmental, and epigenetic factors is involved in the pathogenesis of MS. With the advancement of GWAS, various variants associated with MS have been identified. This study aimed to evaluate the association of single-nucleotide polymorphisms (SNPs) rs4925166 and rs1979277 in the SHMT1, MAZ rs34286592, ERG rs2836425, and L3MBTL3 rs4364506 with MS. In this case-control study, the association of five SNPs in SHMT1, MAZ, ERG, and L3MBTL3 genes with relapsing-remitting MS (RR-MS) was investigated in 190 patients and 200 healthy individuals. Four SNPs including SHMT1 rs4925166, SHMT1 rs1979277, MAZ rs34286592, and L3MBTL3 rs4364506 were genotyped using PCR-RFLP and genotyping of ERG rs2836425 was performed by tetra-primer ARMS PCR. Our findings showed a significant difference in the allelic frequencies for the four SNPs of SHMT1 rs4925166, SHMT1 rs1979277, MAZ rs34286592, and ERG rs2836425, while there were no differences in the allele and genotype frequencies for L3MBTL3 rs4364506. These significant associations were observed for the following genotypes: TT and GG genotypes of SHMT1 rs4925166 (OR 0.47 and 1.90, respectively) genotype GG of SHMT1 rs1979277 (OR 0.63), genotype GG of MAZ rs34286592 (OR 0.61), TC and CC genotypes of ERG rs2836425 (OR 1.89 and 0.50, respectively). Our study highlighted that people who are carrying genotypes including GG (SHMT1 rs4925166) and TC (ERG rs2836425) have the highest susceptibility chance for MS, respectively. However, genotypes TT (SHMT1 rs4925166), CC (ERG rs2836425), GG (MAZ rs34286592), and GG (SHMT1 rs1979277) had the highest negative association (protective effect) with MS, respectively. L3MBTL3 rs4364506 was found neither as a predisposing nor a protective variant.
Collapse
|
24
|
The Myb-related protein MYPOP is a novel intrinsic host restriction factor of oncogenic human papillomaviruses. Oncogene 2018; 37:6275-6284. [PMID: 30018400 PMCID: PMC6265261 DOI: 10.1038/s41388-018-0398-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 06/08/2018] [Accepted: 06/08/2018] [Indexed: 11/08/2022]
Abstract
The skin represents a physical and chemical barrier against invading pathogens, which is additionally supported by restriction factors that provide intrinsic cellular immunity. These factors detect viruses to block their replication cycle. Here, we uncover the Myb-related transcription factor, partner of profilin (MYPOP) as a novel antiviral protein. It is highly expressed in the epithelium and binds to the minor capsid protein L2 and the DNA of human papillomaviruses (HPV), which are the primary causative agents of cervical cancer and other tumors. The early promoter activity and early gene expression of the oncogenic HPV types 16 and 18 is potently silenced by MYPOP. Cellular MYPOP-depletion relieves the restriction of HPV16 infection, demonstrating that MYPOP acts as a restriction factor. Interestingly, we found that MYPOP protein levels are significantly reduced in diverse HPV-transformed cell lines and in HPV-induced cervical cancer. Decades ago it became clear that the early oncoproteins E6 and E7 cooperate to immortalize keratinocytes by promoting degradation of tumor suppressor proteins. Our findings suggest that E7 stimulates MYPOP degradation. Moreover, overexpression of MYPOP blocks colony formation of HPV and non-virally transformed keratinocytes, suggesting that MYPOP exhibits tumor suppressor properties.
Collapse
|
25
|
Chen Z, Stelekati E, Kurachi M, Yu S, Cai Z, Manne S, Khan O, Yang X, Wherry EJ. miR-150 Regulates Memory CD8 T Cell Differentiation via c-Myb. Cell Rep 2018; 20:2584-2597. [PMID: 28903040 DOI: 10.1016/j.celrep.2017.08.060] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2017] [Revised: 06/09/2017] [Accepted: 08/01/2017] [Indexed: 12/22/2022] Open
Abstract
MicroRNAs play an important role in T cell responses. However, how microRNAs regulate CD8 T cell memory remains poorly defined. Here, we found that miR-150 negatively regulates CD8 T cell memory in vivo. Genetic deletion of miR-150 disrupted the balance between memory precursor and terminal effector CD8 T cells following acute viral infection. Moreover, miR-150-deficient memory CD8 T cells were more protective upon rechallenge. A key circuit whereby miR-150 repressed memory CD8 T cell development through the transcription factor c-Myb was identified. Without miR-150, c-Myb was upregulated and anti-apoptotic targets of c-Myb, such as Bcl-2 and Bcl-xL, were also increased, suggesting a miR-150-c-Myb survival circuit during memory CD8 T cell development. Indeed, overexpression of non-repressible c-Myb rescued the memory CD8 T cell defects caused by overexpression of miR-150. Overall, these results identify a key role for miR-150 in memory CD8 T cells through a c-Myb-controlled enhanced survival circuit.
Collapse
Affiliation(s)
- Zeyu Chen
- Department of Microbiology, University of Pennsylvania, Philadelphia, PA, USA; Institute for Immunology, University of Pennsylvania, Philadelphia, PA, USA
| | - Erietta Stelekati
- Department of Microbiology, University of Pennsylvania, Philadelphia, PA, USA; Institute for Immunology, University of Pennsylvania, Philadelphia, PA, USA
| | - Makoto Kurachi
- Department of Microbiology, University of Pennsylvania, Philadelphia, PA, USA; Institute for Immunology, University of Pennsylvania, Philadelphia, PA, USA
| | - Sixiang Yu
- Department of Cancer Biology and Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Zhangying Cai
- Department of Microbiology, University of Pennsylvania, Philadelphia, PA, USA; Institute for Immunology, University of Pennsylvania, Philadelphia, PA, USA; College of Life Sciences, Peking University, Beijing, China
| | - Sasikanth Manne
- Department of Microbiology, University of Pennsylvania, Philadelphia, PA, USA; Institute for Immunology, University of Pennsylvania, Philadelphia, PA, USA
| | - Omar Khan
- Department of Microbiology, University of Pennsylvania, Philadelphia, PA, USA; Institute for Immunology, University of Pennsylvania, Philadelphia, PA, USA
| | - Xiaolu Yang
- Department of Cancer Biology and Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - E John Wherry
- Department of Microbiology, University of Pennsylvania, Philadelphia, PA, USA; Institute for Immunology, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
26
|
Frech M, Teichler S, Feld C, Bouchard C, Berberich H, Sorg K, Mernberger M, Bullinger L, Bauer UM, Neubauer A. MYB induces the expression of the oncogenic corepressor SKI in acute myeloid leukemia. Oncotarget 2018; 9:22423-22435. [PMID: 29854289 PMCID: PMC5976475 DOI: 10.18632/oncotarget.25051] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Accepted: 03/21/2018] [Indexed: 11/25/2022] Open
Abstract
Acute myeloid leukemia (AML) arises through clonal expansion of transformed myeloid progenitor cells. The SKI proto-oncogene is highly upregulated in different solid tumors and leukemic cells, but little is known about its transcriptional regulation during leukemogenesis. MYB is an important hematopoietic transcription factor involved in proliferation as well as differentiation and upregulated in most human acute leukemias. Here, we find that MYB protein binds within the regulatory region of the SKI gene in AML cells. Reporter gene assays using MYB binding sites present in the SKI gene locus show MYB-dependent transcriptional activation. SiRNA-mediated depletion of MYB in leukemic cell lines reveals that MYB is crucial for SKI gene expression. Consistently, we observed a positive correlation of MYB and SKI expression in leukemic cell lines and in samples of AML patients. Moreover, MYB and SKI both were downregulated by treatment with histone deacetylase inhibitors. Strikingly, differentiation of AML cells induced by depletion of MYB is attenuated by overexpression of SKI. Our findings identify SKI as a novel MYB target gene, relevant for the MYB-induced differentiation block in leukemic cells.
Collapse
Affiliation(s)
- Miriam Frech
- Department of Internal Medicine and Hematology, Oncology and Immunology, Philipps University Marburg, Marburg 35033, Germany.,University Hospital Giessen and Marburg, Marburg 35033, Germany
| | - Sabine Teichler
- Department of Internal Medicine and Hematology, Oncology and Immunology, Philipps University Marburg, Marburg 35033, Germany.,University Hospital Giessen and Marburg, Marburg 35033, Germany
| | - Christine Feld
- Department of Internal Medicine and Hematology, Oncology and Immunology, Philipps University Marburg, Marburg 35033, Germany.,University Hospital Giessen and Marburg, Marburg 35033, Germany.,Institute of Molecular Biology and Tumor Research (IMT), School of Medicine, Philipps University Marburg, Marburg 35043, Germany
| | - Caroline Bouchard
- Institute of Molecular Biology and Tumor Research (IMT), School of Medicine, Philipps University Marburg, Marburg 35043, Germany
| | - Hannah Berberich
- Institute of Molecular Biology and Tumor Research (IMT), School of Medicine, Philipps University Marburg, Marburg 35043, Germany
| | - Katharina Sorg
- Department of Internal Medicine and Hematology, Oncology and Immunology, Philipps University Marburg, Marburg 35033, Germany.,University Hospital Giessen and Marburg, Marburg 35033, Germany
| | - Marco Mernberger
- Institute of Molecular Oncology, Philipps University Marburg, Marburg 35043, Germany
| | - Lars Bullinger
- Department of Internal Medicine III, University Hospital of Ulm, Ulm 89081, Germany
| | - Uta-Maria Bauer
- Institute of Molecular Biology and Tumor Research (IMT), School of Medicine, Philipps University Marburg, Marburg 35043, Germany
| | - Andreas Neubauer
- Department of Internal Medicine and Hematology, Oncology and Immunology, Philipps University Marburg, Marburg 35033, Germany.,University Hospital Giessen and Marburg, Marburg 35033, Germany
| |
Collapse
|
27
|
Fahl SP, Daamen AR, Crittenden RB, Bender TP. c-Myb Coordinates Survival and the Expression of Genes That Are Critical for the Pre-BCR Checkpoint. THE JOURNAL OF IMMUNOLOGY 2018; 200:3450-3463. [PMID: 29654210 DOI: 10.4049/jimmunol.1302303] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2013] [Accepted: 03/13/2018] [Indexed: 11/19/2022]
Abstract
The c-Myb transcription factor is required for adult hematopoiesis, yet little is known about c-Myb function during lineage-specific differentiation due to the embryonic lethality of Myb-null mutations. We previously used tissue-specific inactivation of the murine Myb locus to demonstrate that c-Myb is required for differentiation to the pro-B cell stage, survival during the pro-B cell stage, and the pro-B to pre-B cell transition during B lymphopoiesis. However, few downstream mediators of c-Myb-regulated function have been identified. We demonstrate that c-Myb regulates the intrinsic survival of CD19+ pro-B cells in the absence of IL-7 by repressing expression of the proapoptotic proteins Bmf and Bim and that levels of Bmf and Bim mRNA are further repressed by IL-7 signaling in pro-B cells. c-Myb regulates two crucial components of the IL-7 signaling pathway: the IL-7Rα-chain and the negative regulator SOCS3 in CD19+ pro-B cells. Bypassing IL-7R signaling through constitutive activation of Stat5b largely rescues survival of c-Myb-deficient pro-B cells, whereas constitutively active Akt is much less effective. However, rescue of pro-B cell survival is not sufficient to rescue proliferation of pro-B cells or the pro-B to small pre-B cell transition, and we further demonstrate that c-Myb-deficient large pre-B cells are hypoproliferative. Analysis of genes crucial for the pre-BCR checkpoint demonstrates that, in addition to IL-7Rα, the genes encoding λ5, cyclin D3, and CXCR4 are downregulated in the absence of c-Myb, and λ5 is a direct c-Myb target. Thus, c-Myb coordinates survival with the expression of genes that are required during the pre-BCR checkpoint.
Collapse
Affiliation(s)
- Shawn P Fahl
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia Health System, Charlottesville, VA 22908; and
| | - Andrea R Daamen
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia Health System, Charlottesville, VA 22908; and
| | - Rowena B Crittenden
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia Health System, Charlottesville, VA 22908; and
| | - Timothy P Bender
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia Health System, Charlottesville, VA 22908; and .,Beirne B. Carter Center for Immunology Research, University of Virginia Health System, Charlottesville, VA 22908
| |
Collapse
|
28
|
Liu X, Xu Y, Han L, Yi Y. Reassessing the Potential of Myb-targeted Anti-cancer Therapy. J Cancer 2018; 9:1259-1266. [PMID: 29675107 PMCID: PMC5907674 DOI: 10.7150/jca.23992] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Accepted: 02/28/2018] [Indexed: 01/27/2023] Open
Abstract
Transcription factor MYB is essential for the tumorigenesis of multiple cancers, especially leukemia, breast cancer, colon cancer, adenoid cystic carcinoma and brain cancer. Thus, MYB has been regarded as an attractive target for tumor therapy. However, pioneer studies of antisense oligodeoxynucleotides against MYB, which were launched three decades ago in leukemia therapy, were discontinued because of their unsatisfactory clinical outcomes. In recent years, the roles of MYB in tumor transformation have become increasingly clear. Moreover, the regulatory mechanisms of MYB, such as the vital effects of MYB co-regulators on MYB activity and of transcriptional elongation on MYB expression, have been unveiled. These observations have underpinned novel approaches in inhibiting MYB. This review discusses the structure, function and regulation of MYB, focusing on recent insights into MYB-associated oncogenesis and how MYB-targeted therapeutics can be explored. Additionally, the main MYB-targeted therapies, including novel genetic therapy, RNA interference, microRNAs and low-molecular-weight compounds, which are especially promising inhibitors that target MYB co-regulators and transcriptional elongation, are described, and their prospects are assessed.
Collapse
Affiliation(s)
- Xiaofeng Liu
- Department of Hematology, the Second Xiangya Hospital, Central South University, Changsha, Hunan Province, P.R. China
| | - Yunxiao Xu
- Department of Hematology, the Second Xiangya Hospital, Central South University, Changsha, Hunan Province, P.R. China
| | - Liping Han
- School of Life Science, Changchun Normal University, Changchun, Jilin Province, P.R. China
| | - Yan Yi
- Department of Hematology, the Second Xiangya Hospital, Central South University, Changsha, Hunan Province, P.R. China
| |
Collapse
|
29
|
Frerich CA, Brayer KJ, Painter BM, Kang H, Mitani Y, El-Naggar AK, Ness SA. Transcriptomes define distinct subgroups of salivary gland adenoid cystic carcinoma with different driver mutations and outcomes. Oncotarget 2017; 9:7341-7358. [PMID: 29484115 PMCID: PMC5800907 DOI: 10.18632/oncotarget.23641] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Accepted: 12/08/2017] [Indexed: 12/30/2022] Open
Abstract
The relative rarity of salivary gland adenoid cystic carcinoma (ACC) and its slow growing yet aggressive nature has complicated the development of molecular markers for patient stratification. To analyze molecular differences linked to the protracted disease course of ACC and metastases that form 5 or more years after diagnosis, detailed RNA-sequencing (RNA-seq) analysis was performed on 68 ACC tumor samples, starting with archived, formalin-fixed paraffin-embedded (FFPE) samples up to 25 years old, so that clinical outcomes were available. A statistical peak-finding approach was used to classify the tumors that expressed MYB or MYBL1, which had overlapping gene expression signatures, from a group that expressed neither oncogene and displayed a unique phenotype. Expression of MYB or MYBL1 was closely correlated to the expression of the SOX4 and EN1 genes, suggesting that they are direct targets of Myb proteins in ACC tumors. Unsupervised hierarchical clustering identified a subgroup of approximately 20% of patients with exceptionally poor overall survival (median less than 30 months) and a unique gene expression signature resembling embryonic stem cells. The results provide a strategy for stratifying ACC patients and identifying the high-risk, poor-outcome group that are candidates for personalized therapies.
Collapse
Affiliation(s)
- Candace A Frerich
- Department of Internal Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| | - Kathryn J Brayer
- Department of Internal Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM, USA.,University of New Mexico Comprehensive Cancer Center, Albuquerque, NM, USA
| | - Brandon M Painter
- Department of Internal Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| | - Huining Kang
- Department of Internal Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| | - Yoshitsugu Mitani
- Head and Neck Pathology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Adel K El-Naggar
- Head and Neck Pathology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Scott A Ness
- Department of Internal Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM, USA.,University of New Mexico Comprehensive Cancer Center, Albuquerque, NM, USA
| |
Collapse
|
30
|
Álvaro-Blanco J, Urso K, Chiodo Y, Martín-Cortázar C, Kourani O, Arco PGD, Rodríguez-Martínez M, Calonge E, Alcamí J, Redondo JM, Iglesias T, Campanero MR. MAZ induces MYB expression during the exit from quiescence via the E2F site in the MYB promoter. Nucleic Acids Res 2017; 45:9960-9975. [PMID: 28973440 PMCID: PMC5622404 DOI: 10.1093/nar/gkx641] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Accepted: 07/13/2017] [Indexed: 12/27/2022] Open
Abstract
Most E2F-binding sites repress transcription through the recruitment of Retinoblastoma (RB) family members until the end of the G1 cell-cycle phase. Although the MYB promoter contains an E2F-binding site, its transcription is activated shortly after the exit from quiescence, before RB family members inactivation, by unknown mechanisms. We had previously uncovered a nuclear factor distinct from E2F, Myb-sp, whose DNA-binding site overlapped the E2F element and had hypothesized that this factor might overcome the transcriptional repression of MYB by E2F-RB family members. We have purified Myb-sp and discovered that Myc-associated zinc finger proteins (MAZ) are major components. We show that various MAZ isoforms are present in Myb-sp and activate transcription via the MYB-E2F element. Moreover, while forced RB or p130 expression repressed the activity of a luciferase reporter driven by the MYB-E2F element, co-expression of MAZ proteins not only reverted repression, but also activated transcription. Finally, we show that MAZ binds the MYB promoter in vivo, that its binding site is critical for MYB transactivation, and that MAZ knockdown inhibits MYB expression during the exit from quiescence. Together, these data indicate that MAZ is essential to bypass MYB promoter repression by RB family members and to induce MYB expression.
Collapse
Affiliation(s)
- Josué Álvaro-Blanco
- Department of Cancer Biology, Instituto de Investigaciones Biomédicas Alberto Sols, CSIC-UAM, Madrid 28029, Spain
| | - Katia Urso
- Gene regulation in cardiovascular remodeling and inflammation group, Centro Nacional de Investigaciones Cardiovasculares, Madrid 28029, Spain
| | - Yuri Chiodo
- Department of Cancer Biology, Instituto de Investigaciones Biomédicas Alberto Sols, CSIC-UAM, Madrid 28029, Spain
| | - Carla Martín-Cortázar
- Department of Cancer Biology, Instituto de Investigaciones Biomédicas Alberto Sols, CSIC-UAM, Madrid 28029, Spain
| | - Omar Kourani
- Department of Cancer Biology, Instituto de Investigaciones Biomédicas Alberto Sols, CSIC-UAM, Madrid 28029, Spain
| | - Pablo Gómez-Del Arco
- Gene regulation in cardiovascular remodeling and inflammation group, Centro Nacional de Investigaciones Cardiovasculares, Madrid 28029, Spain.,Department of Molecular Biology, Universidad Autónoma de Madrid, Centro de Biología Molecular, Cantoblanco, Madrid 28049, Spain.,CIBERCV, Spain
| | - María Rodríguez-Martínez
- Department of Cancer Biology, Instituto de Investigaciones Biomédicas Alberto Sols, CSIC-UAM, Madrid 28029, Spain
| | - Esther Calonge
- Unidad de Inmunopatología del SIDA, Centro Nacional de Microbiología, Majadahonda 28220, Spain
| | - José Alcamí
- Unidad de Inmunopatología del SIDA, Centro Nacional de Microbiología, Majadahonda 28220, Spain
| | - Juan Miguel Redondo
- Gene regulation in cardiovascular remodeling and inflammation group, Centro Nacional de Investigaciones Cardiovasculares, Madrid 28029, Spain.,CIBERCV, Spain
| | - Teresa Iglesias
- Department of Endocrine and Nervous Systems Pathophysiology, Instituto de Investigaciones Biomédicas Alberto Sols, CSIC-UAM, Madrid 28029, Spain.,CIBERNED, Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas, Spain
| | - Miguel R Campanero
- Department of Cancer Biology, Instituto de Investigaciones Biomédicas Alberto Sols, CSIC-UAM, Madrid 28029, Spain.,CIBERCV, Spain
| |
Collapse
|
31
|
Ness SA. Editorial: Targeting MYB Oncogene Expression in Adenoid Cystic Carcinoma. J Natl Cancer Inst 2017; 109:3845955. [PMID: 28954283 DOI: 10.1093/jnci/djx054] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Accepted: 03/01/2017] [Indexed: 12/11/2022] Open
Affiliation(s)
- Scott A Ness
- Department of Internal Medicine/Molecular Medicine and UNM Comprehensive Cancer Center, University of New Mexico Health Sciences Center, Albuquerque, NM
| |
Collapse
|
32
|
Fujii K, Murase T, Beppu S, Saida K, Takino H, Masaki A, Ijichi K, Kusafuka K, Iida Y, Onitsuka T, Yatabe Y, Hanai N, Hasegawa Y, Inagaki H. MYB,MYBL1,MYBL2andNFIBgene alterations and MYC overexpression in salivary gland adenoid cystic carcinoma. Histopathology 2017; 71:823-834. [DOI: 10.1111/his.13281] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Accepted: 06/05/2017] [Indexed: 12/17/2022]
Affiliation(s)
- Kana Fujii
- Department of Pathology and Molecular Diagnostics; Graduate School of Medical Sciences; Nagoya City University; Nagoya Japan
| | - Takayuki Murase
- Department of Pathology and Molecular Diagnostics; Graduate School of Medical Sciences; Nagoya City University; Nagoya Japan
| | - Shintaro Beppu
- Department of Pathology and Molecular Diagnostics; Graduate School of Medical Sciences; Nagoya City University; Nagoya Japan
- Department of Otolaryngology, Head and Neck Surgery; Graduate School of Medical Sciences; Nagoya City University; Nagoya Japan
| | - Kosuke Saida
- Department of Pathology and Molecular Diagnostics; Graduate School of Medical Sciences; Nagoya City University; Nagoya Japan
| | - Hisashi Takino
- Department of Pathology and Molecular Diagnostics; Graduate School of Medical Sciences; Nagoya City University; Nagoya Japan
| | - Ayako Masaki
- Department of Pathology and Molecular Diagnostics; Graduate School of Medical Sciences; Nagoya City University; Nagoya Japan
| | - Kei Ijichi
- Department of Otolaryngology, Head and Neck Surgery; Graduate School of Medical Sciences; Nagoya City University; Nagoya Japan
| | - Kimihide Kusafuka
- Pathology Division; Shizuoka Cancer Center; Nagaizumi, Shizuoka Japan
| | - Yoshiyuki Iida
- Department of Head and Neck Surgery; Shizuoka Cancer Center; Nagaizumi, Shizuoka Japan
| | - Tetsuro Onitsuka
- Department of Head and Neck Surgery; Shizuoka Cancer Center; Nagaizumi, Shizuoka Japan
| | - Yasushi Yatabe
- Department of Pathology and Molecular Diagnostics; Aichi Cancer Center Hospital; Nagoya Japan
| | - Nobuhiro Hanai
- Department of Head and Neck Surgery; Aichi Cancer Center Hospital; Nagoya Japan
| | - Yasuhisa Hasegawa
- Department of Head and Neck Surgery; Aichi Cancer Center Hospital; Nagoya Japan
| | - Hiroshi Inagaki
- Department of Pathology and Molecular Diagnostics; Graduate School of Medical Sciences; Nagoya City University; Nagoya Japan
| |
Collapse
|
33
|
Ruvolo PP. GSK-3 as a novel prognostic indicator in leukemia. Adv Biol Regul 2017; 65:26-35. [PMID: 28499784 DOI: 10.1016/j.jbior.2017.05.001] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Revised: 05/05/2017] [Accepted: 05/07/2017] [Indexed: 06/07/2023]
Abstract
While leukemias represent a diverse set of diseases with malignant cells derived from myeloid or lymphoid origin, a common feature is the dysregulation of signal transduction pathways that influence leukemogeneisis, promote drug resistance, and favor leukemia stem cells. Mutations in PI3K, PTEN, RAS, or other upstream regulators can activate the AKT kinase which has central roles in supporting cell proliferation and survival. A major target of AKT is Glycogen Synthase Kinase 3 (GSK3). GSK3 has two isoforms (alpha and beta) that were studied as regulators of metabolism but emerged as central players in cancer in the early 1990s. GSK3 is unique in that the isoforms are constitutively active. Active GSK3 promotes destruction of oncogenic proteins such as beta Catenin, c-MYC, and MCL-1 and thus has tumor suppressor properties. In AML, inactivation of GSK3 is associated with poor overall survival. Interestingly in some leukemias GSK3 targets a tumor suppressor and thus the kinases can act as tumor promoters in those instances. An example is GSK3 targeting p27Kip1 in AML with MLL translocation. This review will cover the role of GSK3 in various leukemias both as tumor suppressor and tumor promoter. We will also briefly cover current state of GSK3 inhibitors for leukemia therapy.
Collapse
Affiliation(s)
- Peter P Ruvolo
- Department of Leukemia, Unit 448, The University of Texas M.D. Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, United States.
| |
Collapse
|
34
|
Kim D, You E, Jeong J, Ko P, Kim JW, Rhee S. DDR2 controls the epithelial-mesenchymal-transition-related gene expression via c-Myb acetylation upon matrix stiffening. Sci Rep 2017; 7:6847. [PMID: 28754957 PMCID: PMC5533734 DOI: 10.1038/s41598-017-07126-7] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Accepted: 06/23/2017] [Indexed: 02/03/2023] Open
Abstract
Increasing matrix stiffness caused by the extracellular matrix (ECM) deposition surrounding cancer cells is accompanied by epithelial-mesenchymal transition (EMT). Here, we show that expression levels of EMT marker genes along with discoidin domain receptor 2 (DDR2) can increase upon matrix stiffening. DDR2 silencing by short hairpin RNA downregulated EMT markers. Promoter analysis and chromatin immunoprecipitation revealed that c-Myb and LEF1 may be responsible for DDR2 induction during cell culture on a stiff matrix. Mechanistically, c-Myb acetylation by p300, which is upregulated on the stiff matrix, seems to be necessary for the c-Myb-and-LEF1-mediated DDR2 expression. Finally, we found that the c-Myb-DDR2 axis is crucial for lung cancer cell line proliferation and expression of EMT marker genes in a stiff environment. Thus, our results suggest that DDR2 regulation by p300 expression and/or c-Myb acetylation upon matrix stiffening may be necessary for regulation of EMT and invasiveness of lung cancer cells.
Collapse
Affiliation(s)
- Daehwan Kim
- Department of Life Science, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Eunae You
- Department of Life Science, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Jangho Jeong
- Department of Life Science, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Panseon Ko
- Department of Life Science, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Jung-Woong Kim
- Department of Life Science, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Sangmyung Rhee
- Department of Life Science, Chung-Ang University, Seoul, 06974, Republic of Korea.
| |
Collapse
|
35
|
Brown RB, Madrid NJ, Suzuki H, Ness SA. Optimized approach for Ion Proton RNA sequencing reveals details of RNA splicing and editing features of the transcriptome. PLoS One 2017; 12:e0176675. [PMID: 28459821 PMCID: PMC5411089 DOI: 10.1371/journal.pone.0176675] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Accepted: 04/14/2017] [Indexed: 11/29/2022] Open
Abstract
RNA-sequencing (RNA-seq) has become the standard method for unbiased analysis of gene expression but also provides access to more complex transcriptome features, including alternative RNA splicing, RNA editing, and even detection of fusion transcripts formed through chromosomal translocations. However, differences in library methods can adversely affect the ability to recover these different types of transcriptome data. For example, some methods have bias for one end of transcripts or rely on low-efficiency steps that limit the complexity of the resulting library, making detection of rare transcripts less likely. We tested several commonly used methods of RNA-seq library preparation and found vast differences in the detection of advanced transcriptome features, such as alternatively spliced isoforms and RNA editing sites. By comparing several different protocols available for the Ion Proton sequencer and by utilizing detailed bioinformatics analysis tools, we were able to develop an optimized random primer based RNA-seq technique that is reliable at uncovering rare transcript isoforms and RNA editing features, as well as fusion reads from oncogenic chromosome rearrangements. The combination of optimized libraries and rapid Ion Proton sequencing provides a powerful platform for the transcriptome analysis of research and clinical samples.
Collapse
Affiliation(s)
- Roger B. Brown
- Department of Internal Medicine, Division of Molecular Medicine, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, United States of America
| | - Nathaniel J. Madrid
- Department of Internal Medicine, Division of Molecular Medicine, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, United States of America
| | - Hideaki Suzuki
- Department of Internal Medicine, Division of Molecular Medicine, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, United States of America
| | - Scott A. Ness
- Department of Internal Medicine, Division of Molecular Medicine, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, United States of America
- UNM Comprehensive Cancer Center, University of New Mexico, Albuquerque, New Mexico, United States of America
- * E-mail:
| |
Collapse
|
36
|
Chi K, Li Y, Xu L, Wang X. A novel recurrent copy number loss region on 6q23.3 in MDS-related myeloid malignancy patients with stable survival conditions. Leuk Lymphoma 2017; 58:2470-2479. [PMID: 28394181 DOI: 10.1080/10428194.2017.1292357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Metaphase cytogenetics (MC) karyotyping is a fundamental way to approach cytogenetic pathogenesis of MDS-related myeloid malignancies. However, in some patients, the results are normal while the patients often show discrepancies in survival conditions. To explain this question, we analyzed CytoScan™ HD array results of 20 MC-normal/failure patients who were followed up for three years. Exon sequencing was performed in genes RUNX1, TP53, ASXL1, and TET2. The array enabled the detection of additional aberrations in 16 (80%) patients. Eight patients were detected with cryptic copy number losses and six of them got aggressive disease conditions. RUNX1 mutations were sequenced in P110 and P114. Most importantly, two patients (P114 and P116) with copy number loss aberrations got stable survival conditions during follow-ups, and a novel recurrent copy number loss region harboring the proto-oncogene MYB was detected on chromosome 6q23.3 in both of them, which might benefit the survival of the patients.
Collapse
Affiliation(s)
- Kun Chi
- a Department of Laboratory Medicine , Qingdao Women & Children's Hospital , Qingdao , China.,b State Key Laboratory of Medical Genomics , Shanghai Institute of Hematology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine , Shanghai , China
| | - Yang Li
- b State Key Laboratory of Medical Genomics , Shanghai Institute of Hematology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine , Shanghai , China
| | - Lan Xu
- c Department of Hematology , Ruijin Hospital, Shanghai Jiaotong University School of Medicine , Shanghai , China
| | - Xuefeng Wang
- d Department of Laboratory Medicine , Ruijin Hospital, Shanghai Jiaotong University School of Medicine , Shanghai , China
| |
Collapse
|
37
|
Lu J, Zhang M, Yang X, Cui T, Dai J. MicroRNA-548c-3p inhibits T98G glioma cell proliferation and migration by downregulating c-Myb. Oncol Lett 2017; 13:3866-3872. [PMID: 28536644 DOI: 10.3892/ol.2017.5870] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Accepted: 01/10/2017] [Indexed: 01/03/2023] Open
Abstract
MicroRNAs (miRNAs/miRs) are short non-coding RNAs (between 20 and 22 nucleotides) that regulate gene expression by binding to the 3'-untranslated region of target mRNA, and preventing protein translation or inducing mRNA destabilization. miRNAs are predicted to target ~60% of all mRNAs, therefore providing a marked degree of regulation of a number of cellular processes. In the present study, the expression of miR-548c-3p was determined by reverse transcription-quantitative polymerase chain reaction analysis and demonstrated to be markedly downregulated in clinical malignant glioma tissues and the glioma T98G cell line compared with normal human brain tissue. Transfection of miR-548c-3p inhibited cell proliferation by inducing G1 cell cycle arrest and also inhibited the migration of the T98G cells in vitro. Furthermore, a bioinformatic algorithm and a luciferase reporter assay identified proto-oncogene c-Myb (c-Myb) as a potential direct target of miR-548c-3p. Further experiments demonstrated that the inhibition of c-Myb by miR-548c-3p partially mediated the antitumor effect of miR-548c-3p. The results of the present study provide the novel insight that miR-548c-3p inhibits glioma tumorigenesis by targeting c-Myb. Therefore, miR-548c-3p may contribute to the development of improved glioma treatment.
Collapse
Affiliation(s)
- Jianyi Lu
- Department of Medical Genetics and Developmental Biology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, P.R. China.,School of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325027, P.R. China
| | - Min Zhang
- School of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325027, P.R. China
| | - Xiao Yang
- School of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325027, P.R. China
| | - Tong Cui
- School of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325027, P.R. China
| | - Jinpo Dai
- School of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325027, P.R. China.,State Key Laboratory Cultivation Base and Key Laboratory of Vision Science, Ministry of Health of The People's Republic of China, Zhejiang Provincial Key Laboratory of Ophthalmology and Optometry, Wenzhou, Zhejiang 325027, P.R. China
| |
Collapse
|
38
|
Guo H, Zhang B, Nairn AV, Nagy T, Moremen KW, Buckhaults P, Pierce M. O-Linked N-Acetylglucosamine ( O-GlcNAc) Expression Levels Epigenetically Regulate Colon Cancer Tumorigenesis by Affecting the Cancer Stem Cell Compartment via Modulating Expression of Transcriptional Factor MYBL1. J Biol Chem 2017; 292:4123-4137. [PMID: 28096468 DOI: 10.1074/jbc.m116.763201] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Revised: 01/15/2017] [Indexed: 12/19/2022] Open
Abstract
To study the regulation of colorectal adenocarcinoma progression by O-GlcNAc, we have focused on the O-GlcNAc-mediated epigenetic regulation of human colon cancer stem cells (CCSC). Xenograft tumors from colon tumor cells with O-linked N-acetylglucosamine transferase (OGT) knockdown grew significantly slower than those formed from control cells, indicating a reduced proliferation of tumor cells due to inhibition of OGT expression. Significant reduction of the CCSC population was observed in the tumor cells after OGT knockdown, whereas tumor cells treated with the O-GlcNAcase inhibitor showed an increased CCSC population, indicating that O-GlcNAc levels regulated the CCSC compartment. When grown in suspension, tumor cells with OGT knockdown showed a reduced ability to form tumorspheres, indicating a reduced self-renewal of CCSC due to reduced levels of O-GlcNAc. ChIP-sequencing experiments using an anti-O-GlcNAc antibody revealed significant chromatin enrichment of O-GlcNAc-modified proteins at the promoter of the transcription factor MYBL1, which was also characterized by the presence of H3K27me3. RNA-sequencing analysis showed an increased expression of MYBL1 in tumor cells with OGT knockdown. Forced overexpression of MYBL1 led to a reduced population of CCSC and tumor growth in vivo, similar to the effects of OGT silencing. Moreover, two CpG islands near the transcription start site of MYBL1 were identified, and O-GlcNAc levels regulated their methylation status. These results strongly argue that O-GlcNAc epigenetically regulates MYBL1, functioning similarly to H3K27me3. The aberrant CCSC compartment observed after modulating O-GlcNAc levels is therefore likely to result, at least in part, from the epigenetic regulation of MYBL1 expression by O-GlcNAc, thereby significantly affecting tumor progression.
Collapse
Affiliation(s)
- Huabei Guo
- From the Department of Biochemistry and Molecular Biology, Complex Carbohydrate Research Center, and
| | - Bing Zhang
- the Boston Children's Hospital, Harvard University, Boston, Massachusetts 02115, and
| | - Alison V Nairn
- From the Department of Biochemistry and Molecular Biology, Complex Carbohydrate Research Center, and
| | - Tamas Nagy
- Department of Pathology, College of Veterinary Medicine, University of Georgia, Athens, Georgia 30602
| | - Kelley W Moremen
- From the Department of Biochemistry and Molecular Biology, Complex Carbohydrate Research Center, and
| | - Phillip Buckhaults
- the South Carolina College of Pharmacy, University of South Carolina, Columbia, South Carolina 29208
| | - Michael Pierce
- From the Department of Biochemistry and Molecular Biology, Complex Carbohydrate Research Center, and
| |
Collapse
|
39
|
Uttarkar S, Frampton J, Klempnauer KH. Targeting the transcription factor Myb by small-molecule inhibitors. Exp Hematol 2016; 47:31-35. [PMID: 28017646 DOI: 10.1016/j.exphem.2016.12.003] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Accepted: 12/10/2016] [Indexed: 11/18/2022]
Abstract
The transcription factor Myb is a key regulator of hematopoietic cell proliferation, differentiation, and survival and has been implicated in the development of leukemia and several other human cancers. Pharmacological inhibition of Myb is therefore emerging as a potential therapeutic strategy. Recently, the first low-molecular-weight compounds that show Myb inhibitory activity have been identified. Characterization of these compounds suggests disruption of the protein-protein-interaction of Myb and the coactivator p300 as a suitable strategy to inhibit Myb.
Collapse
Affiliation(s)
| | - Jon Frampton
- Institute of Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham, United Kingdom
| | | |
Collapse
|
40
|
Fernandes Q. MicroRNA: Defining a new niche in Leukemia. Blood Rev 2016; 31:129-138. [PMID: 28087197 DOI: 10.1016/j.blre.2016.11.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Revised: 09/10/2016] [Accepted: 11/22/2016] [Indexed: 12/19/2022]
Abstract
MicroRNAs (miRNAs) are endogenous short non-coding RNAs found to play key roles in the pathogenesis of leukemia. Apart from being traditionally identified as modulators of oncogenes, the potential roles of miRNAs seems to be growing with novel and recent findings among different subtypes of hematological malignancies. Leukemia is one of the earliest malignancies to be linked to abnormal expression of miRNAs. However, a clear understanding of the involvement of miRNAs in intricate mechanisms of leukemogenesis is still a necessity. This review summarizes the multiple roles of miRNAs in the pathogenesis of leukemia and highlights major research findings contributing to these aspects.
Collapse
|
41
|
Yu L, Ham K, Gao X, Castro L, Yan Y, Kissling GE, Tucker CJ, Flagler N, Dong R, Archer TK, Dixon D. Epigenetic regulation of transcription factor promoter regions by low-dose genistein through mitogen-activated protein kinase and mitogen-and-stress activated kinase 1 nongenomic signaling. Cell Commun Signal 2016; 14:18. [PMID: 27582276 PMCID: PMC5007815 DOI: 10.1186/s12964-016-0141-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Accepted: 08/17/2016] [Indexed: 02/06/2023] Open
Abstract
Background The phytoestrogen, genistein at low doses nongenomically activates mitogen-activated protein kinase p44/42 (MAPKp44/42) via estrogen receptor alpha (ERα) leading to proliferation of human uterine leiomyoma cells. In this study, we evaluated if MAPKp44/42 could activate downstream effectors such as mitogen- and stress-activated protein kinase 1 (MSK1), which could then epigenetically modify histone H3 by phosphorylation following a low dose (1 μg/ml) of genistein. Results Using hormone-responsive immortalized human uterine leiomyoma (ht-UtLM) cells, we found that genistein activated MAPKp44/42 and MSK1, and also increased phosphorylation of histone H3 at serine10 (H3S10ph) in ht-UtLM cells. Colocalization of phosphorylated MSK1 and H3S10ph was evident by confocal microscopy in ht-UtLM cells (r = 0.8533). Phosphorylation of both MSK1and H3S10ph was abrogated by PD98059 (PD), a MEK1 kinase inhibitor, thereby supporting genistein’s activation of MSK1 and Histone H3 was downstream of MAPKp44/42. In proliferative (estrogenic) phase human uterine fibroid tissues, phosphorylated MSK1 and H3S10ph showed increased immunoexpression compared to normal myometrial tissues, similar to results observed in in vitro studies following low-dose genistein administration. Real-time RT-PCR arrays showed induction of growth-related transcription factor genes, EGR1, Elk1, ID1, and MYB (cMyb) with confirmation by western blot, downstream of MAPK in response to low-dose genistein in ht-UtLM cells. Additionally, genistein induced associations of promoter regions of the above transcription factors with H3S10ph as evidenced by Chromatin Immunoprecipitation (ChIP) assays, which were inhibited by PD. Therefore, genistein epigenetically modified histone H3 by phosphorylation of serine 10, which was regulated by MSK1 and MAPK activation. Conclusion Histone H3 phosphorylation possibly represents a mechanism whereby increased transcriptional activation occurs following low-dose genistein exposure. Electronic supplementary material The online version of this article (doi:10.1186/s12964-016-0141-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Linda Yu
- Molecular Pathogenesis Group, National Toxicology Program (NTP) Laboratory, Division of the NTP (DNTP), National Institute of Environmental Health Sciences (NIEHS), National Institutes of Health (NIH), U.S. Department of Health and Human Services (HHS), Research Triangle Park, North Carolina, 27709, USA
| | - Kyle Ham
- Molecular Pathogenesis Group, National Toxicology Program (NTP) Laboratory, Division of the NTP (DNTP), National Institute of Environmental Health Sciences (NIEHS), National Institutes of Health (NIH), U.S. Department of Health and Human Services (HHS), Research Triangle Park, North Carolina, 27709, USA
| | - Xiaohua Gao
- Molecular Pathogenesis Group, National Toxicology Program (NTP) Laboratory, Division of the NTP (DNTP), National Institute of Environmental Health Sciences (NIEHS), National Institutes of Health (NIH), U.S. Department of Health and Human Services (HHS), Research Triangle Park, North Carolina, 27709, USA
| | - Lysandra Castro
- Molecular Pathogenesis Group, National Toxicology Program (NTP) Laboratory, Division of the NTP (DNTP), National Institute of Environmental Health Sciences (NIEHS), National Institutes of Health (NIH), U.S. Department of Health and Human Services (HHS), Research Triangle Park, North Carolina, 27709, USA
| | - Yitang Yan
- Molecular Pathogenesis Group, National Toxicology Program (NTP) Laboratory, Division of the NTP (DNTP), National Institute of Environmental Health Sciences (NIEHS), National Institutes of Health (NIH), U.S. Department of Health and Human Services (HHS), Research Triangle Park, North Carolina, 27709, USA
| | - Grace E Kissling
- Biostatistics and Computational Biology Branch, Division of Intramural Research (DIR), NIEHS, NIH, HHS, Research Triangle Park, North Carolina, 27709, USA
| | - Charles J Tucker
- Signal Transduction Laboratory, DIR, NIEHS, NIH, HHS, Research Triangle Park, North Carolina, 27709, USA
| | - Norris Flagler
- Cellular and Molecular Pathology Branch, DNTP, NIEHS, NIH, HHS, Research Triangle Park, North Carolina, 27709, USA
| | - Ray Dong
- Molecular Pathogenesis Group, National Toxicology Program (NTP) Laboratory, Division of the NTP (DNTP), National Institute of Environmental Health Sciences (NIEHS), National Institutes of Health (NIH), U.S. Department of Health and Human Services (HHS), Research Triangle Park, North Carolina, 27709, USA
| | - Trevor K Archer
- Chromatin and Gene Expression Group, Epigenetics and Stem Cell Biology Laboratory, DIR, NIEHS, NIH, HHS, Research Triangle Park, North Carolina, 27709, USA
| | - Darlene Dixon
- Molecular Pathogenesis Group, National Toxicology Program (NTP) Laboratory, Division of the NTP (DNTP), National Institute of Environmental Health Sciences (NIEHS), National Institutes of Health (NIH), U.S. Department of Health and Human Services (HHS), Research Triangle Park, North Carolina, 27709, USA.
| |
Collapse
|
42
|
Yarbrough WG, Panaccione A, Chang MT, Ivanov SV. Clinical and molecular insights into adenoid cystic carcinoma: Neural crest-like stemness as a target. Laryngoscope Investig Otolaryngol 2016; 1:60-77. [PMID: 28894804 PMCID: PMC5510248 DOI: 10.1002/lio2.22] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Accepted: 05/10/2016] [Indexed: 12/16/2022] Open
Abstract
OBJECTIVES This review surveys trialed therapies and molecular defects in adenoid cystic carcinoma (ACC), with an emphasis on neural crest-like stemness characteristics of newly discovered cancer stem cells (CSCs) and therapies that may target these CSCs. DATA SOURCES Articles available on Pubmed or OVID MEDLINE databases and unpublished data. REVIEW METHODS Systematic review of articles pertaining to ACC and neural crest-like stem cells. RESULTS Adenoid cystic carcinoma of the salivary gland is a slowly growing but relentless cancer that is prone to nerve invasion and metastases. A lack of understanding of molecular etiology and absence of targetable drivers has limited therapy for patients with ACC to surgery and radiation. Currently, no curative treatments are available for patients with metastatic disease, which highlights the need for effective new therapies. Research in this area has been inhibited by the lack of validated cell lines and a paucity of clinically useful markers. The ACC research environment has recently improved, thanks to the introduction of novel tools, technologies, approaches, and models. Improved understanding of ACC suggests that neural crest-like stemness is a major target in this rare tumor. New cell culture techniques and patient-derived xenografts provide tools for preclinical testing. CONCLUSION Preclinical research has not identified effective targets in ACC, as confirmed by the large number of failed clinical trials. New molecular data suggest that drivers of neural crest-like stemness may be required for maintenance of ACC; as such, CSCs are a target for therapy of ACC.
Collapse
Affiliation(s)
- Wendell G. Yarbrough
- Section of Otolaryngology, Department of Surgery, Yale School of MedicineNew HavenConnecticutUSA
- Yale Cancer CenterNew HavenConnecticutUSA
| | - Alexander Panaccione
- Department of Cancer BiologyVanderbilt University School of MedicineNashvilleTennesseeU.S.A.
| | - Michael T. Chang
- Section of Otolaryngology, Department of Surgery, Yale School of MedicineNew HavenConnecticutUSA
| | - Sergey V. Ivanov
- Section of Otolaryngology, Department of Surgery, Yale School of MedicineNew HavenConnecticutUSA
| |
Collapse
|
43
|
Corda G, Sala A. Cutaneous cylindroma: it's all about MYB. J Pathol 2016; 239:391-3. [PMID: 27185061 DOI: 10.1002/path.4746] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Accepted: 05/11/2016] [Indexed: 12/30/2022]
Abstract
Cutaneous cylindroma is a rare benign tumour that occasionally turns into malignant cylindrocarcinoma. The cancer can be sporadic or emerge in the context of Brooke-Spiegler syndrome (BSS), an inheritable condition characterized by mutation of the gene CYLD, encoding a tumour suppressor protein that controls the activity of the transcription factor NF-kB. Sporadic cylindromas present histological features shared with adenoid cystic carcinoma (ACC), a head and neck cancer originating from salivary or other exocrine glands. Like ACCs, sporadic cylindromas express, although at lower frequency, the aberrant fusion transcript MYB-NFIB. In a paper recently published in the Journal of Pathology, the research teams led by Neil Rajan and Goran Stenman demonstrate that CYLD-defective cyclindromas in BSS patients are negative for the MYB-NFIB fusion. Only the wild-type MYB oncoprotein is activated in the majority of these tumours. RNA interference studies in cells derived from BSS patients indicate that ablating MYB expression results in a striking reduction of cylindroma cell proliferation, suggesting that MYB plays a pivotal role in the biology of this cancer. The take-home message of the study is that activation of MYB, in its wild-type form or fusion derivatives, is a common feature of spontaneous and hereditary cylindromas, constituting a potentially actionable therapeutic target. Copyright © 2016 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Gabriele Corda
- College of Health and Life Sciences, Brunel University, London, UK.,Institute of Environment, Health and Societies, Brunel University, London, UK
| | - Arturo Sala
- College of Health and Life Sciences, Brunel University, London, UK.,Institute of Environment, Health and Societies, Brunel University, London, UK
| |
Collapse
|
44
|
PIAS1 binds p300 and behaves as a coactivator or corepressor of the transcription factor c-Myb dependent on SUMO-status. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2016; 1859:705-18. [PMID: 27032383 DOI: 10.1016/j.bbagrm.2016.03.011] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Revised: 03/08/2016] [Accepted: 03/23/2016] [Indexed: 12/21/2022]
Abstract
The PIAS proteins (Protein Inhibitor of Activated STATs) constitute a family of multifunctional nuclear proteins operating as SUMO E3 ligases and being involved in a multitude of interactions. They participate in a range of biological processes, also beyond their well-established role in the immune system and cytokine signalling. They act both as transcriptional corepressors and coactivators depending on the context. In the present work, we investigated mechanisms by which PIAS1 causes activation or repression of c-Myb dependent target genes. Analysis of global expression data shows that c-Myb and PIAS1 knockdowns affect a subset of common targets, but with a dual outcome consistent with a role of PIAS1 as either a corepressor or coactivator. Our mechanistic studies show that PIAS1 engages in a novel interaction with the acetyltransferase and coactivator p300. Interaction and ChIP analysis suggest a bridging function where PIAS1 enhances p300 recruitment to c-Myb-bound sites through interaction with both proteins. In addition, the E3 activity of PIAS1 enhances further its coactivation. Remarkably, the SUMO status of c-Myb had a decisive role, indicating a SUMO-dependent switch in the way PIAS1 affects c-Myb, either as a coactivator or corepressor. Removal of the two major SUMO-conjugation sites in c-Myb (2KR mutant), which enhances its activity significantly, turned PIAS1 into a corepressor. Also, p300 was less efficiently recruited to chromatin by c-Myb-2KR. We propose that PIAS1 acts as a "protein inhibitor of activated c-Myb" in the absence of SUMOylation while, in its presence, PIAS behaves as a "protein activator of repressed c-Myb".
Collapse
|
45
|
Inoue K, Fry EA. Novel Molecular Markers for Breast Cancer. BIOMARKERS IN CANCER 2016; 8:25-42. [PMID: 26997872 PMCID: PMC4790586 DOI: 10.4137/bic.s38394] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/30/2015] [Revised: 02/16/2016] [Accepted: 02/14/2016] [Indexed: 01/15/2023]
Abstract
The use of molecular biomarkers assures that breast cancer (BC) patients receive optimal treatment. Established biomarkers, such as estrogen receptor, progesterone receptor, HER2, and Ki67, have been playing significant roles in the subcategorization of BC to predict the prognosis and decide the specific therapy to each patient. Antihormonal therapy using 4-hydroxytamoxifen or aromatase inhibitors have been employed in patients whose tumor cells express hormone receptors, while monoclonal antibody to HER2 has been administered to HER2-positive BCs. Although new therapeutic agents have been developed in the past few decades, many patients still die of the disease due to relapse; thus, novel molecular markers that predict therapeutic failure and those that can be targets for specific therapy are expected. We have chosen four of such molecules by reviewing recent publications, which are cyclin E, B-Myb, Twist, and DMP1β. The oncogenicity of these molecules has been demonstrated in vivo and/or in vitro through studies using transgenic mice or siRNAs, and their expressions have been shown to be associated with shortened overall or disease-free survival of BC patients. The former three molecules have been shown to accelerate epithelial-mesenchymal transition that is often associated with cancer stem cell-ness and metastasis; all these four can be novel therapeutic targets as well. Thus, large prospective studies employing immunohistochemistry will be needed to establish the predictive values of these molecules in patients with BC.
Collapse
Affiliation(s)
- Kazushi Inoue
- Department of Pathology, Wake Forest University Health Sciences, Medical Center Boulevard, Winston-Salem, NC, USA
| | - Elizabeth A. Fry
- Department of Pathology, Wake Forest University Health Sciences, Medical Center Boulevard, Winston-Salem, NC, USA
| |
Collapse
|
46
|
Corcoran LM, Tarlinton DM. Regulation of germinal center responses, memory B cells and plasma cell formation-an update. Curr Opin Immunol 2016; 39:59-67. [PMID: 26799208 DOI: 10.1016/j.coi.2015.12.008] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Revised: 12/21/2015] [Accepted: 12/31/2015] [Indexed: 12/31/2022]
Abstract
Progress in understanding humoral immunity has been accelerated by the powerful experimental approaches of genetics, genomics and imaging. Excellent reviews of these advances appeared in 2015 in celebration of the 50th anniversary of the discovery of B cell and T cell lineages in the chicken. Here we provide a contemporary model of B cell differentiation, highlighting recent publications illuminating germinal center (GC), memory B cell and antibody-secreting plasma cell biology. The important contributions of CD4T cells to antibody responses have been thoroughly reviewed elsewhere.
Collapse
Affiliation(s)
- Lynn M Corcoran
- Molecular Immunology Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia; Department of Medical Biology, The University of Melbourne, Parkville, Victoria 3052, Australia.
| | - David M Tarlinton
- Immunology Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia; Department of Medical Biology, The University of Melbourne, Parkville, Victoria 3052, Australia
| |
Collapse
|
47
|
Guo X, Zhang J, Pang J, He S, Li G, Chong Y, Li C, Jiao Z, Zhang S, Shao M. MicroRNA-503 represses epithelial–mesenchymal transition and inhibits metastasis of osteosarcoma by targeting c-myb. Tumour Biol 2016; 37:9181-7. [DOI: 10.1007/s13277-016-4797-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2015] [Accepted: 01/06/2016] [Indexed: 12/25/2022] Open
|
48
|
Brayer KJ, Frerich CA, Kang H, Ness SA. Recurrent Fusions in MYB and MYBL1 Define a Common, Transcription Factor-Driven Oncogenic Pathway in Salivary Gland Adenoid Cystic Carcinoma. Cancer Discov 2015; 6:176-87. [PMID: 26631070 DOI: 10.1158/2159-8290.cd-15-0859] [Citation(s) in RCA: 162] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Accepted: 11/24/2015] [Indexed: 01/10/2023]
Abstract
UNLABELLED Adenoid cystic carcinoma (ACC), the second most common malignancy of salivary glands, is a rare tumor with a bleak prognosis for which therapeutic targets are unavailable. We used RNA sequencing (RNA-seq) to analyze low-quality RNA from archival, formaldehyde-fixed, paraffin-embedded samples. In addition to detecting the most common ACC translocation, t(6;9) fusing the MYB proto-oncogene to NFIB, we also detected previously unknown t(8;9) and t(8;14) translocations fusing the MYBL1 gene to the NFIB and RAD51B genes, respectively. RNA-seq provided information about gene fusions, alternative RNA splicing, and gene expression signatures. Interestingly, tumors with MYB and MYBL1 translocations displayed similar gene expression profiles, and the combined MYB and MYBL1 expression correlated with outcome, suggesting that the related MYB proteins are interchangeable oncogenic drivers in ACC. Our results provide important details about the biology of ACC and illustrate how archival tissue samples can be used for detailed molecular analyses of rare tumors. SIGNIFICANCE Using RNA-seq to perform whole-transcriptome analysis of archival ACC tumor samples, we identified novel, recurrent gene fusions, detected alternative RNA splicing, and established gene expression signatures that provide detailed information about the biology of ACC tumors.
Collapse
Affiliation(s)
- Kathryn J Brayer
- Department of Internal Medicine and UNM Comprehensive Cancer Center, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| | - Candace A Frerich
- Department of Internal Medicine and UNM Comprehensive Cancer Center, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| | - Huining Kang
- Department of Internal Medicine and UNM Comprehensive Cancer Center, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| | - Scott A Ness
- Department of Internal Medicine and UNM Comprehensive Cancer Center, University of New Mexico Health Sciences Center, Albuquerque, New Mexico.
| |
Collapse
|
49
|
Kaspar P, Zikova M, Bartunek P, Sterba J, Strnad H, Kren L, Sedlacek R. The Expression of c-Myb Correlates with the Levels of Rhabdomyosarcoma-specific Marker Myogenin. Sci Rep 2015; 5:15090. [PMID: 26462877 PMCID: PMC4604482 DOI: 10.1038/srep15090] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2015] [Accepted: 09/14/2015] [Indexed: 12/29/2022] Open
Abstract
The transcription factor c-Myb is required for modulation of progenitor cells in several tissues, including skeletal muscle and its upregulation is observed in many human malignancies. Rhabdomyosarcomas (RMS) are a heterogeneous group of mesodermal tumors with features of developing skeletal muscle. Several miRNAs are downregulated in RMS, including miR-150, a negative regulator of c-Myb expression. Using the C2C12 myoblast cell line, a cellular model of skeletal muscle differentiation, we showed that miR-150 controls c-Myb expression mainly at the level of translation. We hypothesized that a similar mechanism of c-Myb regulation operates in RMS tumors. We examined expression of c-Myb by immunohistochemistry and revealed c-Myb positivity in alveolar and embryonal tumors, the two most common subgroups of RMS. Furthermore, we showed direct correlation between c-Myb production and myogenin expression. Interestingly, high myogenin levels indicate poor prognosis in RMS patients. c-Myb could, therefore, contribute to the tumor phenotype by executing its inhibitory role in skeletal muscle differentiation. We also showed that c-Myb protein is abundant in migratory C2C12 myoblasts and its ectopic expression potentiates cell motility. In summary, our results implicate that metastatic properties of some RMS subtypes might be linked to c-Myb function.
Collapse
Affiliation(s)
- Petr Kaspar
- Laboratory of Transgenic Models of Diseases, Institute of Molecular Genetics of the ASCR, v.v.i., Prague, Czech Republic
| | - Martina Zikova
- Laboratory of Cell Differentiation, Institute of Molecular Genetics of the ASCR, v.v.i., Prague, Czech Republic
| | - Petr Bartunek
- Laboratory of Cell Differentiation, Institute of Molecular Genetics of the ASCR, v.v.i., Prague, Czech Republic
| | | | - Hynek Strnad
- Laboratory of Genomics and Bioinformatics, Institute of Molecular Genetics of the ASCR, v.v.i., Prague, Czech Republic
| | - Leos Kren
- The University Hospital Brno, Brno, Czech Republic
| | - Radislav Sedlacek
- Laboratory of Transgenic Models of Diseases, Institute of Molecular Genetics of the ASCR, v.v.i., Prague, Czech Republic
| |
Collapse
|
50
|
Hoareau-Aveilla C, Valentin T, Daugrois C, Quelen C, Mitou G, Quentin S, Jia J, Spicuglia S, Ferrier P, Ceccon M, Giuriato S, Gambacorti-Passerini C, Brousset P, Lamant L, Meggetto F. Reversal of microRNA-150 silencing disadvantages crizotinib-resistant NPM-ALK(+) cell growth. J Clin Invest 2015; 125:3505-18. [PMID: 26258416 DOI: 10.1172/jci78488] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2014] [Accepted: 06/23/2015] [Indexed: 01/20/2023] Open
Abstract
The regulatory microRNA miR-150 is involved in the development of hemopathies and is downregulated in T-lymphomas, such as anaplastic large-cell lymphoma (ALCL) tumors. ALCL is defined by the presence or absence of translocations that activate the anaplastic lymphoma kinase (ALK), with nucleophosmin-ALK (NPM-ALK) fusions being the most common. Here, we compared samples of primary NPM-ALK(+) and NPM-ALK(-) ALCL to investigate the role of miR-150 downstream of NPM-ALK. Methylation of the MIR150 gene was substantially elevated in NPM-ALK(+) biopsies and correlated with reduced miR-150 expression. In NPM-ALK(+) cell lines, DNA hypermethylation-mediated miR-150 repression required ALK-dependent pathways, as ALK inhibition restored miR-150 expression. Moreover, epigenetic silencing of miR-150 was due to the activation of STAT3, a major downstream substrate of NPM-ALK, in cooperation with DNA methyltransferase 1 (DNMT1). Accordingly, miR-150 repression was turned off following treatment with the DNMT inhibitor, decitabine. In murine NPM-ALK(+) xenograft models, miR-150 upregulation induced antineoplastic activity. Treatment of crizotinib-resistant NPM-ALK(+) KARPAS-299-CR06 cells with decitabine or ectopic miR-150 expression reduced viability and growth. Altogether, our results suggest that hypomethylating drugs, alone or in combination with other agents, may benefit ALK(+) patients harboring tumors resistant to crizotinib and other anti-ALK tyrosine kinase inhibitors (TKIs). Moreover, these results support further work on miR-150 in these and other ALK(+) malignancies.
Collapse
MESH Headings
- Animals
- Cell Line, Tumor
- Crizotinib
- DNA (Cytosine-5-)-Methyltransferase 1
- DNA (Cytosine-5-)-Methyltransferases/genetics
- DNA (Cytosine-5-)-Methyltransferases/metabolism
- Drug Resistance, Neoplasm
- Female
- Gene Expression Regulation, Neoplastic
- Gene Silencing
- Humans
- Lymphoma, Large-Cell, Anaplastic/drug therapy
- Lymphoma, Large-Cell, Anaplastic/genetics
- Lymphoma, Large-Cell, Anaplastic/metabolism
- Lymphoma, Large-Cell, Anaplastic/pathology
- Male
- Mice
- Mice, Transgenic
- MicroRNAs/biosynthesis
- MicroRNAs/genetics
- Protein-Tyrosine Kinases/genetics
- Protein-Tyrosine Kinases/metabolism
- Pyrazoles/pharmacology
- Pyridines/pharmacology
- RNA, Neoplasm/biosynthesis
- RNA, Neoplasm/genetics
- STAT3 Transcription Factor/genetics
- STAT3 Transcription Factor/metabolism
Collapse
|