1
|
Fliorent R, Hoda ST, Jour G, Mantilla JG. Spindle Cell Sarcoma With Novel JAZF1::NUDT5 Gene Fusion: Report of a Previously Undescribed Neoplasm. Genes Chromosomes Cancer 2025; 64:e70026. [PMID: 39873241 DOI: 10.1002/gcc.70026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 01/06/2025] [Accepted: 01/15/2025] [Indexed: 01/30/2025] Open
Abstract
Gene fusions involving JAZF1 are a recurrent event in low grade endometrial stromal sarcoma, and have been more recently described in few instances of endometrial stromal sarcoma-like tumors in the genitourinary tract of men. In this article, we describe a previously unreported spindle cell sarcoma harboring an in-frame JAZF1::NUDT5 gene fusion, arising in the chest wall of a 51-year-old man. The tumor had unique morphologic features resembling both endometrial stromal sarcoma and endometrial stromal sarcoma-like tumors, consisting of a mixture of cytologically bland and pleomorphic spindle cells with brisk mitotic activity, within an alternating myxoid and fibrous stroma. It had diffuse immunohistochemical expression of CD10, CD34 and CD56, and variable expression of androgen receptor. To our knowledge, neoplasms with these clinico-pathologic characteristics and novel gene fusion have not been previously reported in the English language literature.
Collapse
Affiliation(s)
- Rebecca Fliorent
- Rowan-Virtua School of Osteopathic Medicine, Stratford, New Jersey, USA
| | - Syed T Hoda
- Department of Pathology, NYU Grossman School of Medicine, New York, New York, USA
| | - George Jour
- Department of Pathology, NYU Grossman School of Medicine, New York, New York, USA
| | - Jose G Mantilla
- Department of Pathology, NYU Grossman School of Medicine, New York, New York, USA
| |
Collapse
|
2
|
Alkafaas SS, Khedr SA, ElKafas SS, Hafez W, Loutfy SA, Sakran M, Janković N. Targeting JNK kinase inhibitors via molecular docking: A promising strategy to address tumorigenesis and drug resistance. Bioorg Chem 2024; 153:107776. [PMID: 39276490 DOI: 10.1016/j.bioorg.2024.107776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 07/13/2024] [Accepted: 08/28/2024] [Indexed: 09/17/2024]
Abstract
Among members of the mitogen-activated protein kinase (MAPK) family, c-Jun N-terminal kinases (JNKs) are vital for cellular responses to stress, inflammation, and apoptosis. Recent advances have highlighted their important implications in cancer biology, where dysregulated JNK signalling plays a role in the growth, progression, and metastasis of tumors. The present understanding of JNK kinase and its function in the etiology of cancer is summarized in this review. By modifying a number of downstream targets, such as transcription factors, apoptotic regulators, and cell cycle proteins, JNKs exert diverse effects on cancer cells. Apoptosis avoidance, cell survival, and proliferation are all promoted by abnormal JNK activation in many types of cancer, which leads to tumor growth and resistance to treatment. JNKs also affect the tumour microenvironment by controlling the generation of inflammatory cytokines, angiogenesis, and immune cell activity. However, challenges remain in deciphering the context-specific roles of JNK isoforms and their intricate crosstalk with other signalling pathways within the complex tumor environment. Further research is warranted to delineate the precise mechanisms underlying JNK-mediated tumorigenesis and to develop tailored therapeutic strategies targeting JNK signalling to improve cancer management. The review emphasizes the role of JNK kinases in cancer biology, as well as their potential as pharmaceutical targets for precision oncology therapy and cancer resistance. Also, this review summarizes all the available promising JNK inhibitors that are suggested to promote the responsiveness of cancer cells to cancer treatment.
Collapse
Affiliation(s)
- Samar Sami Alkafaas
- Molecular Cell Biology Unit, Division of Biochemistry, Department of Chemistry, Faculty of Science, Tanta University, 31527, Egypt.
| | - Sohila A Khedr
- Industrial Biotechnology Department, Faculty of Science, Tanta University, Tanta 31733, Egypt
| | - Sara Samy ElKafas
- Production Engineering and Mechanical Design Department, Faculty of Engineering, Menofia University, Menofia, Egypt; Faculty of Control System and Robotics, ITMO University, Saint-Petersburg, Russia
| | - Wael Hafez
- NMC Royal Hospital, 16th St - Khalifa City - SE-4 - Abu Dhabi, United Arab Emirates; Department of Internal Medicine, Medical Research and Clinical Studies Institute, The National Research Centre, 33 El Buhouth St, Ad Doqi, Dokki, Cairo Governorate 12622, Egypt
| | - Samah A Loutfy
- Virology and Immunology Unit, Cancer Biology Department, National Cancer Institute, Cairo University, Cairo, Egypt
| | - Mohamed Sakran
- Biochemistry Department, Faculty of Science, University of Tabuk, Tabuk 47512, Saudi Arabia
| | - Nenad Janković
- Institute for Information Technologies Kragujevac, Department of Science, University of Kragujevac, Jovana Cvijića bb, 34000 Kragujevac, Serbia.
| |
Collapse
|
3
|
Chen R, Tang L, Melendy T, Yang L, Goodison S, Sun Y. Prostate Cancer Progression Modeling Provides Insight into Dynamic Molecular Changes Associated with Progressive Disease States. CANCER RESEARCH COMMUNICATIONS 2024; 4:2783-2798. [PMID: 39347576 PMCID: PMC11500312 DOI: 10.1158/2767-9764.crc-24-0210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Revised: 08/27/2024] [Accepted: 09/25/2024] [Indexed: 10/01/2024]
Abstract
Prostate cancer is a significant health concern and the most commonly diagnosed cancer in men worldwide. Understanding the complex process of prostate tumor evolution and progression is crucial for improved diagnosis, treatments, and patient outcomes. Previous studies have focused on unraveling the dynamics of prostate cancer evolution using phylogenetic or lineage analysis approaches. However, those approaches have limitations in capturing the complete disease process or incorporating genomic and transcriptomic variations comprehensively. In this study, we applied a novel computational approach to derive a prostate cancer progression model using multidimensional data from 497 prostate tumor samples and 52 tumor-adjacent normal samples obtained from The Cancer Genome Atlas study. The model was validated using data from an independent cohort of 545 primary tumor samples. By integrating transcriptomic and genomic data, our model provides a comprehensive view of prostate tumor progression, identifies crucial signaling pathways and genetic events, and uncovers distinct transcription signatures associated with disease progression. Our findings have significant implications for cancer research and hold promise for guiding personalized treatment strategies in prostate cancer. SIGNIFICANCE We developed and validated a progression model of prostate cancer using >1,000 tumor and normal tissue samples. The model provided a comprehensive view of prostate tumor evolution and progression.
Collapse
Affiliation(s)
- Runpu Chen
- Department of Microbiology and Immunology, University at Buffalo, State University of New York, Buffalo, New York
| | - Li Tang
- Department of Cancer Prevention and Control, Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | - Thomas Melendy
- Department of Microbiology and Immunology, University at Buffalo, State University of New York, Buffalo, New York
| | - Le Yang
- Department of Microbiology and Immunology, University at Buffalo, State University of New York, Buffalo, New York
| | - Steve Goodison
- Department of Quantitative Health Sciences, Mayo Clinic, Jacksonville, Florida
| | - Yijun Sun
- Department of Microbiology and Immunology, University at Buffalo, State University of New York, Buffalo, New York
- Department of Computer Science and Engineering, University at Buffalo, State University of New York, Buffalo, New York
| |
Collapse
|
4
|
Yan H, He L, Lv D, Yang J, Yuan Z. The Role of the Dysregulated JNK Signaling Pathway in the Pathogenesis of Human Diseases and Its Potential Therapeutic Strategies: A Comprehensive Review. Biomolecules 2024; 14:243. [PMID: 38397480 PMCID: PMC10887252 DOI: 10.3390/biom14020243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 02/12/2024] [Accepted: 02/15/2024] [Indexed: 02/25/2024] Open
Abstract
JNK is named after c-Jun N-terminal kinase, as it is responsible for phosphorylating c-Jun. As a member of the mitogen-activated protein kinase (MAPK) family, JNK is also known as stress-activated kinase (SAPK) because it can be activated by extracellular stresses including growth factor, UV irradiation, and virus infection. Functionally, JNK regulates various cell behaviors such as cell differentiation, proliferation, survival, and metabolic reprogramming. Dysregulated JNK signaling contributes to several types of human diseases. Although the role of the JNK pathway in a single disease has been summarized in several previous publications, a comprehensive review of its role in multiple kinds of human diseases is missing. In this review, we begin by introducing the landmark discoveries, structures, tissue expression, and activation mechanisms of the JNK pathway. Next, we come to the focus of this work: a comprehensive summary of the role of the deregulated JNK pathway in multiple kinds of diseases. Beyond that, we also discuss the current strategies for targeting the JNK pathway for therapeutic intervention and summarize the application of JNK inhibitors as well as several challenges now faced. We expect that this review can provide a more comprehensive insight into the critical role of the JNK pathway in the pathogenesis of human diseases and hope that it also provides important clues for ameliorating disease conditions.
Collapse
Affiliation(s)
- Huaying Yan
- Department of Ultrasound, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China; (H.Y.); (L.H.)
| | - Lanfang He
- Department of Ultrasound, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China; (H.Y.); (L.H.)
| | - De Lv
- Department of Endocrinology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
| | - Jun Yang
- Cancer Center and State Key Laboratory of Biotherapy, Department of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China;
| | - Zhu Yuan
- Cancer Center and State Key Laboratory of Biotherapy, Department of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China;
| |
Collapse
|
5
|
Tai YT, Fukuda T, Morozumi Y, Hirai H, Oda AH, Kamada Y, Akikusa Y, Kanki T, Ohta K, Shiozaki K. Fission Yeast TORC1 Promotes Cell Proliferation through Sfp1, a Transcription Factor Involved in Ribosome Biogenesis. Mol Cell Biol 2023; 43:675-692. [PMID: 38051102 PMCID: PMC10761059 DOI: 10.1080/10985549.2023.2282349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 10/29/2023] [Accepted: 11/06/2023] [Indexed: 12/07/2023] Open
Abstract
Target of rapamycin complex 1 (TORC1) is activated in response to nutrient availability and growth factors, promoting cellular anabolism and proliferation. To explore the mechanism of TORC1-mediated proliferation control, we performed a genetic screen in fission yeast and identified Sfp1, a zinc-finger transcription factor, as a multicopy suppressor of temperature-sensitive TORC1 mutants. Our observations suggest that TORC1 phosphorylates Sfp1 and protects Sfp1 from proteasomal degradation. Transcription analysis revealed that Sfp1 positively regulates genes involved in ribosome production together with two additional transcription factors, Ifh1/Crf1 and Fhl1. Ifh1 physically interacts with Fhl1, and the nuclear localization of Ifh1 is regulated in response to nutrient levels in a manner dependent on TORC1 and Sfp1. Taken together, our data suggest that the transcriptional regulation of the genes involved in ribosome biosynthesis by Sfp1, Ifh1, and Fhl1 is one of the key pathways through which nutrient-activated TORC1 promotes cell proliferation.
Collapse
Affiliation(s)
- Yen Teng Tai
- Division of Biological Science, Nara Institute of Science and Technology, Ikoma, Nara, Japan
| | - Tomoyuki Fukuda
- Department of Cellular Physiology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Yuichi Morozumi
- Division of Biological Science, Nara Institute of Science and Technology, Ikoma, Nara, Japan
| | - Hayato Hirai
- Department of Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo, Tokyo, Japan
| | - Arisa H. Oda
- Department of Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo, Tokyo, Japan
| | - Yoshiaki Kamada
- National Institute for Basic Biology, Okazaki, Aichi, Japan
- Graduate University for Advanced Studies (SOKENDAI), Hayama, Kanagawa, Japan
| | - Yutaka Akikusa
- Division of Biological Science, Nara Institute of Science and Technology, Ikoma, Nara, Japan
| | - Tomotake Kanki
- Department of Cellular Physiology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Kunihiro Ohta
- Department of Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo, Tokyo, Japan
| | - Kazuhiro Shiozaki
- Division of Biological Science, Nara Institute of Science and Technology, Ikoma, Nara, Japan
- Department of Microbiology and Molecular Genetics, University of California, Davis, California, USA
| |
Collapse
|
6
|
Rosario SR, Jacobi JJ, Long MD, Affronti HC, Rowsam AM, Smiraglia DJ. JAZF1: A Metabolic Regulator of Sensitivity to a Polyamine-Targeted Therapy. Mol Cancer Res 2023; 21:24-35. [PMID: 36166196 PMCID: PMC9808368 DOI: 10.1158/1541-7786.mcr-22-0316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 08/05/2022] [Accepted: 09/22/2022] [Indexed: 02/03/2023]
Abstract
Identifying and leveraging unique points of metabolic dysregulation in different disease settings is vital for safe and effective incorporation of metabolism-targeted therapies in the clinic. In addition, it has been shown identification of master metabolic transcriptional regulators (MMTR) of individual metabolic pathways, and how they relate to the disease in question, may offer the key to understanding therapeutic response. In prostate cancer, we have previously demonstrated polyamine biosynthesis and the methionine cycle were targetable metabolic vulnerabilities. However, the MMTRs of these pathways, and how they affect treatment, have yet to be explored. We sought to characterize differential sensitivity of prostate cancer to polyamine- and methionine-targeted therapies by identifying novel MMTRs. We began by developing a gene signature from patient samples, which can predict response to metabolic therapy, and further uncovered a MMTR, JAZF1. We characterized the effects of JAZF1 overexpression on prostate cancer cells, basally and in the context of treatment, by assessing mRNA levels, proliferation, colony formation capability, and key metabolic processes. Lastly, we confirmed the relevance of our findings in large publicly available cohorts of prostate cancer patient samples. We demonstrated differential sensitivity to polyamine and methionine therapies and identified JAZF1 as a MMTR of this response. IMPLICATIONS We have shown JAZF1 can alter sensitivity of cells and its expression can segregate patient populations into those that do, or do not highly express polyamine genes, leading to better prediction of response to a polyamine targeting therapy.
Collapse
Affiliation(s)
- Spencer R. Rosario
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, New York
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | - Justine J. Jacobi
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | - Mark D. Long
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | - Hayley C. Affronti
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | - Aryn M. Rowsam
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | - Dominic J. Smiraglia
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, New York
| |
Collapse
|
7
|
Lee HY, Jang HR, Li H, Samuel VT, Dudek KD, Osipovich AB, Magnuson MA, Sklar J, Shulman GI. Deletion of Jazf1 gene causes early growth retardation and insulin resistance in mice. Proc Natl Acad Sci U S A 2022; 119:e2213628119. [PMID: 36442127 PMCID: PMC9894197 DOI: 10.1073/pnas.2213628119] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Single-nucleotide polymorphisms in the human juxtaposed with another zinc finger protein 1 (JAZF1) gene have repeatedly been associated with both type 2 diabetes (T2D) and height in multiple genome-wide association studies (GWAS); however, the mechanism by which JAZF1 causes these traits is not yet known. To investigate the possible functional role of JAZF1 in growth and glucose metabolism in vivo, we generated Jazf1 knockout (KO) mice and examined body composition and insulin sensitivity both in young and adult mice by using 1H-nuclear magnetic resonance and hyperinsulinemic-euglycemic clamp techniques. Plasma concentrations of insulin-like growth factor 1 (IGF-1) were reduced in both young and adult Jazf1 KO mice, and young Jazf1 KO mice were shorter in stature than age-matched wild-type mice. Young Jazf1 KO mice manifested reduced fat mass, whereas adult Jazf1 KO mice manifested increased fat mass and reductions in lean body mass associated with increased plasma growth hormone (GH) concentrations. Adult Jazf1 KO manifested muscle insulin resistance that was further exacerbated by high-fat diet feeding. Gene set enrichment analysis in Jazf1 KO liver identified the hepatocyte hepatic nuclear factor 4 alpha (HNF4α), which was decreased in Jazf1 KO liver and in JAZF1 knockdown cells. Moreover, GH-induced IGF-1 expression was inhibited by JAZF1 knockdown in human hepatocytes. Taken together these results demonstrate that reduction of JAZF1 leads to early growth retardation and late onset insulin resistance in vivo which may be mediated through alterations in the GH-IGF-1 axis and HNF4α.
Collapse
Affiliation(s)
- Hui-Young Lee
- aLaboratory of Mitochondria and Metabolic Diseases, School of Medicine, Gachon University, Incheon21999, Korea
- bDepartment of Molecular Medicine, School of Medicine, Gachon University, Incheon21999, Korea
- cKorea Mouse Metabolic Phenotyping Center, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon21999, Korea
| | - Hye Rim Jang
- aLaboratory of Mitochondria and Metabolic Diseases, School of Medicine, Gachon University, Incheon21999, Korea
- bDepartment of Molecular Medicine, School of Medicine, Gachon University, Incheon21999, Korea
| | - Hui Li
- dDepartment of Pathology, University of Virginia, Charlottesville, VA22908
| | - Varman T. Samuel
- eDepartment of Internal Medicine, Yale School of Medicine, New Haven, CT06510
- fWest Haven Veterans Affairs Medical Center, West Haven, CT06516
| | - Karrie D. Dudek
- gDepartment of Cell and Developmental Biology, Vanderbilt University, NashvilleTN37232
| | - Anna B. Osipovich
- hDepartment of Molecular Physiology and Biophysics, Vanderbilt UniversityNashville, TN37232
| | - Mark A. Magnuson
- hDepartment of Molecular Physiology and Biophysics, Vanderbilt UniversityNashville, TN37232
- 1To whom correspondence may be addressed. , , or
| | - Jeffrey Sklar
- iDepartment of Pathology, Yale School of Medicine, New HavenCT06510
- 1To whom correspondence may be addressed. , , or
| | - Gerald I. Shulman
- eDepartment of Internal Medicine, Yale School of Medicine, New Haven, CT06510
- jDepartment of Cellular & Molecular Physiology, Yale School of Medicine, New Haven, CT06510
- 1To whom correspondence may be addressed. , , or
| |
Collapse
|
8
|
Sánchez-Maldonado JM, Collado R, Cabrera-Serrano AJ, Ter Horst R, Gálvez-Montosa F, Robles-Fernández I, Arenas-Rodríguez V, Cano-Gutiérrez B, Bakker O, Bravo-Fernández MI, García-Verdejo FJ, López JAL, Olivares-Ruiz J, López-Nevot MÁ, Fernández-Puerta L, Cózar-Olmo JM, Li Y, Netea MG, Jurado M, Lorente JA, Sánchez-Rovira P, Álvarez-Cubero MJ, Sainz J. Type 2 Diabetes-Related Variants Influence the Risk of Developing Prostate Cancer: A Population-Based Case-Control Study and Meta-Analysis. Cancers (Basel) 2022; 14:cancers14102376. [PMID: 35625981 PMCID: PMC9139180 DOI: 10.3390/cancers14102376] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 04/26/2022] [Accepted: 04/29/2022] [Indexed: 02/06/2023] Open
Abstract
In this study, we have evaluated whether 57 genome-wide association studies (GWAS)-identified common variants for type 2 diabetes (T2D) influence the risk of developing prostate cancer (PCa) in a population of 304 Caucasian PCa patients and 686 controls. The association of selected single nucleotide polymorphisms (SNPs) with the risk of PCa was validated through meta-analysis of our data with those from the UKBiobank and FinnGen cohorts, but also previously published genetic studies. We also evaluated whether T2D SNPs associated with PCa risk could influence host immune responses by analysing their correlation with absolute numbers of 91 blood-derived cell populations and circulating levels of 103 immunological proteins and 7 steroid hormones. We also investigated the correlation of the most interesting SNPs with cytokine levels after in vitro stimulation of whole blood, peripheral mononuclear cells (PBMCs), and monocyte-derived macrophages with LPS, PHA, Pam3Cys, and Staphylococcus Aureus. The meta-analysis of our data with those from six large cohorts confirmed that each copy of the FTOrs9939609A, HNF1Brs7501939T, HNF1Brs757210T, HNF1Brs4430796G, and JAZF1rs10486567A alleles significantly decreased risk of developing PCa (p = 3.70 × 10-5, p = 9.39 × 10-54, p = 5.04 × 10-54, p = 1.19 × 10-71, and p = 1.66 × 10-18, respectively). Although it was not statistically significant after correction for multiple testing, we also found that the NOTCH2rs10923931T and RBMS1rs7593730 SNPs associated with the risk of developing PCa (p = 8.49 × 10-4 and 0.004). Interestingly, we found that the protective effect attributed to the HFN1B locus could be mediated by the SULT1A1 protein (p = 0.00030), an arylsulfotransferase that catalyzes the sulfate conjugation of many hormones, neurotransmitters, drugs, and xenobiotic compounds. In addition to these results, eQTL analysis revealed that the HNF1Brs7501939, HNF1Brs757210, HNF1Brs4430796, NOTCH2rs10923931, and RBMS1rs7593730 SNPs influence the risk of PCa through the modulation of mRNA levels of their respective genes in whole blood and/or liver. These results confirm that functional TD2-related variants influence the risk of developing PCa, but also highlight the need of additional experiments to validate our functional results in a tumoral tissue context.
Collapse
Affiliation(s)
- José Manuel Sánchez-Maldonado
- Genomic Oncology Area, GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, PTS Granada, 18016 Granada, Spain; (J.M.S.-M.); (A.J.C.-S.); (I.R.-F.); (V.A.-R.); (M.J.); (J.A.L.); (M.J.Á.-C.)
- Hematology Department, Virgen de las Nieves University Hospital, 18012 Granada, Spain;
- Instituto de Investigación Biosanataria IBs. Granada, 18012 Granada, Spain
| | - Ricardo Collado
- Medical Oncology Department, Hospital de San Pedro Alcántara, 10003 Cáceres, Spain; (R.C.); (M.I.B.-F.); (J.O.-R.)
| | - Antonio José Cabrera-Serrano
- Genomic Oncology Area, GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, PTS Granada, 18016 Granada, Spain; (J.M.S.-M.); (A.J.C.-S.); (I.R.-F.); (V.A.-R.); (M.J.); (J.A.L.); (M.J.Á.-C.)
- Hematology Department, Virgen de las Nieves University Hospital, 18012 Granada, Spain;
- Instituto de Investigación Biosanataria IBs. Granada, 18012 Granada, Spain
| | - Rob Ter Horst
- Department of Internal Medicine and Radboud Centre for Infectious Diseases, Radboud University Nijmegen Medical Center, 6525 GA Nijmegen, The Netherlands; (R.T.H.); (Y.L.); (M.G.N.)
| | - Fernando Gálvez-Montosa
- Department of Medical Oncology, Complejo Hospitalario de Jaén, 23007 Jaén, Spain; (F.G.-M.); (F.J.G.-V.); (J.A.L.L.); (P.S.-R.)
| | - Inmaculada Robles-Fernández
- Genomic Oncology Area, GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, PTS Granada, 18016 Granada, Spain; (J.M.S.-M.); (A.J.C.-S.); (I.R.-F.); (V.A.-R.); (M.J.); (J.A.L.); (M.J.Á.-C.)
| | - Verónica Arenas-Rodríguez
- Genomic Oncology Area, GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, PTS Granada, 18016 Granada, Spain; (J.M.S.-M.); (A.J.C.-S.); (I.R.-F.); (V.A.-R.); (M.J.); (J.A.L.); (M.J.Á.-C.)
- Department of Biochemistry and Molecular Biology III, Faculty of Medicine, University of Granada, 18016 Granada, Spain;
| | - Blanca Cano-Gutiérrez
- Department of Biochemistry and Molecular Biology III, Faculty of Medicine, University of Granada, 18016 Granada, Spain;
| | - Olivier Bakker
- Department of Genetics, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands;
| | | | - Francisco José García-Verdejo
- Department of Medical Oncology, Complejo Hospitalario de Jaén, 23007 Jaén, Spain; (F.G.-M.); (F.J.G.-V.); (J.A.L.L.); (P.S.-R.)
| | - José Antonio López López
- Department of Medical Oncology, Complejo Hospitalario de Jaén, 23007 Jaén, Spain; (F.G.-M.); (F.J.G.-V.); (J.A.L.L.); (P.S.-R.)
| | - Jesús Olivares-Ruiz
- Medical Oncology Department, Hospital de San Pedro Alcántara, 10003 Cáceres, Spain; (R.C.); (M.I.B.-F.); (J.O.-R.)
| | | | | | | | - Yang Li
- Department of Internal Medicine and Radboud Centre for Infectious Diseases, Radboud University Nijmegen Medical Center, 6525 GA Nijmegen, The Netherlands; (R.T.H.); (Y.L.); (M.G.N.)
- Centre for Individualised Infection Medicine (CiiM) & TWINCORE, Joint Ventures between the Helmholtz-Centre for Infection Research (HZI) and the Hannover Medical School (MHH), 30625 Hannover, Germany
| | - Mihai G. Netea
- Department of Internal Medicine and Radboud Centre for Infectious Diseases, Radboud University Nijmegen Medical Center, 6525 GA Nijmegen, The Netherlands; (R.T.H.); (Y.L.); (M.G.N.)
- Department for Immunology & Metabolism, Life and Medical Sciences Institute (LIMES), University of Bonn, 53115 Bonn, Germany
| | - Manuel Jurado
- Genomic Oncology Area, GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, PTS Granada, 18016 Granada, Spain; (J.M.S.-M.); (A.J.C.-S.); (I.R.-F.); (V.A.-R.); (M.J.); (J.A.L.); (M.J.Á.-C.)
- Hematology Department, Virgen de las Nieves University Hospital, 18012 Granada, Spain;
- Instituto de Investigación Biosanataria IBs. Granada, 18012 Granada, Spain
- Department of Medicine, Faculty of Medicine, University of Granada, 18016 Granada, Spain
| | - Jose Antonio Lorente
- Genomic Oncology Area, GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, PTS Granada, 18016 Granada, Spain; (J.M.S.-M.); (A.J.C.-S.); (I.R.-F.); (V.A.-R.); (M.J.); (J.A.L.); (M.J.Á.-C.)
- Department of Legal Medicine, Faculty of Medicine, University of Granada, 18016 Granada, Spain
| | - Pedro Sánchez-Rovira
- Department of Medical Oncology, Complejo Hospitalario de Jaén, 23007 Jaén, Spain; (F.G.-M.); (F.J.G.-V.); (J.A.L.L.); (P.S.-R.)
| | - María Jesús Álvarez-Cubero
- Genomic Oncology Area, GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, PTS Granada, 18016 Granada, Spain; (J.M.S.-M.); (A.J.C.-S.); (I.R.-F.); (V.A.-R.); (M.J.); (J.A.L.); (M.J.Á.-C.)
- Department of Biochemistry and Molecular Biology III, Faculty of Medicine, University of Granada, 18016 Granada, Spain;
| | - Juan Sainz
- Genomic Oncology Area, GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, PTS Granada, 18016 Granada, Spain; (J.M.S.-M.); (A.J.C.-S.); (I.R.-F.); (V.A.-R.); (M.J.); (J.A.L.); (M.J.Á.-C.)
- Hematology Department, Virgen de las Nieves University Hospital, 18012 Granada, Spain;
- Instituto de Investigación Biosanataria IBs. Granada, 18012 Granada, Spain
- Department of Biochemistry and Molecular Biology I, Faculty of Sciences, University of Granada, 18071 Granada, Spain
- Correspondence: ; Tel.: +34-95871-5500 (ext. 126); Fax: +34-9-5863-7071
| |
Collapse
|
9
|
Jeong J, Jang S, Park S, Kwon W, Kim SY, Jang S, Ko J, Park SJ, Lim SG, Yoon D, Yi J, Lee S, Kim MO, Choi SK, Ryoo ZY. JAZF1 heterozygous knockout mice show altered adipose development and metabolism. Cell Biosci 2021; 11:161. [PMID: 34407873 PMCID: PMC8375039 DOI: 10.1186/s13578-021-00625-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Accepted: 06/10/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Juxtaposed with another zinc finger protein 1 (JAZF1) is associated with metabolic disorders, including type 2 diabetes mellitus (T2DM). Several studies showed that JAZF1 and body fat mass are closely related. We attempted to elucidate the JAZF1 functions on adipose development and related metabolism using in vitro and in vivo models. RESULTS The JAZF1 expression was precisely regulated during adipocyte differentiation of 3T3-L1 preadipocyte and mouse embryonic fibroblasts (MEFs). Homozygous JAZF1 deletion (JAZF1-KO) resulted in impaired adipocyte differentiation in MEF. The JAZF1 role in adipocyte differentiation was demonstrated by the regulation of PPARγ-a key regulator of adipocyte differentiation. Heterozygous JAZF1 deletion (JAZF1-Het) mice fed a normal diet (ND) or a high-fat diet (HFD) had less adipose tissue mass and impaired glucose homeostasis than the control (JAZF1-Cont) mice. However, other metabolic organs, such as brown adipose tissue and liver, were negligible effect on JAZF1 deficiency. CONCLUSION Our findings emphasized the JAZF1 role in adipocyte differentiation and related metabolism through the heterozygous knockout mice. This study provides new insights into the JAZF1 function in adipose development and metabolism, informing strategies for treating obesity and related metabolic disorders.
Collapse
Affiliation(s)
- Jain Jeong
- Digestive Diseases Section, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Soyoung Jang
- School of Life Sciences, BK21 FOUR KNU Creative BioResearch, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Song Park
- Core Protein Resources Center, DGIST, Daegu, 42988, Republic of Korea.,Department of Brain and Cognitive Sciences, DGIST, Daegu, Republic of Korea
| | - Wookbong Kwon
- Division of Biotechnology, DGIST, Daegu, Republic of Korea
| | - Si-Yong Kim
- School of Life Sciences, BK21 FOUR KNU Creative BioResearch, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Soyoen Jang
- School of Life Sciences, BK21 FOUR KNU Creative BioResearch, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Jiwon Ko
- School of Life Sciences, BK21 FOUR KNU Creative BioResearch, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Si Jun Park
- School of Life Sciences, BK21 FOUR KNU Creative BioResearch, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Su-Geun Lim
- School of Life Sciences, BK21 FOUR KNU Creative BioResearch, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Duhak Yoon
- Department of Animal Science, Kyungpook National University, Daegu, 37224, Republic of Korea
| | - Junkoo Yi
- Gyeongsangbukdo Livestock Research Institute, Yeongju, Republic of Korea
| | - Sanggyu Lee
- School of Life Sciences, BK21 FOUR KNU Creative BioResearch, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Myoung Ok Kim
- School of Animal Science and Biotechnology, Kyungpook National University, Daegu, Korea
| | - Seong-Kyoon Choi
- Core Protein Resources Center, DGIST, Daegu, 42988, Republic of Korea. .,Division of Biotechnology, DGIST, Daegu, Republic of Korea.
| | - Zae Young Ryoo
- School of Life Sciences, BK21 FOUR KNU Creative BioResearch, Kyungpook National University, Daegu, 41566, Republic of Korea.
| |
Collapse
|
10
|
Li C, Wang C. LG-ESSs and HG-ESSs: underlying molecular alterations and potential therapeutic strategies. J Zhejiang Univ Sci B 2021; 22:633-646. [PMID: 34414699 PMCID: PMC8377580 DOI: 10.1631/jzus.b2000797] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 03/19/2021] [Accepted: 03/19/2021] [Indexed: 12/29/2022]
Abstract
Endometrial stromal tumors (ESTs) include endometrial stromal nodule (ESN), low-grade endometrial stromal sarcoma (LG-ESS), high-grade endometrial stromal sarcoma (HG-ESS), and undifferentiated uterine sarcoma (UUS). Since these are rare tumor types, there is an unmet clinical need for the systematic therapy of advanced LG-ESS or HG-ESS. Cytogenetic and molecular advances in ESTs have shown that multiple recurrent gene fusions are present in a large proportion of LG-ESSs, and HG-ESSs are identified by the tyrosine 3-monooxygenase/tryptophan 5-monooxygenase activation protein epsilon (YWHAE)-family with sequence similarity 22 (FAM22) fusion. Recently, a group of ESSs harboring both zinc finger CCCH domain-containing protein 7B (ZC3H7B)-B-cell lymphoma 6 corepressor(BCOR) fusion and internal tandem duplication (ITD) of the BCOR gene have been provisionally classified as HG-ESSs. In this review, we firstly describe current knowledge about the molecular characteristics of recurrent aberrant proteins and their roles in the tumorigenesis of LG-ESSs and HG-ESSs. Next, we summarize the possibly shared signal pathways in the tumorigenesis of LG-ESSs and HG-ESSs, and list potentially actionable targets. Finally, based on the above discussion, we propose a few promising therapeutic strategies for LG-ESSs and HG-ESSs with recurrent gene alterations.
Collapse
Affiliation(s)
- Chunhui Li
- Quality Management Office, The Second Hospital of Jilin University, Changchun 130041, China
| | - Chunhong Wang
- Department of Hematology and Oncology, The Second Hospital of Jilin University, Changchun 130041, China.
| |
Collapse
|
11
|
Lu B, Jiang R, Xie B, Wu W, Zhao Y. Fusion genes in gynecologic tumors: the occurrence, molecular mechanism and prospect for therapy. Cell Death Dis 2021; 12:783. [PMID: 34381020 PMCID: PMC8357806 DOI: 10.1038/s41419-021-04065-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 07/21/2021] [Accepted: 07/23/2021] [Indexed: 12/12/2022]
Abstract
Gene fusions are thought to be driver mutations in multiple cancers and are an important factor for poor patient prognosis. Most of them appear in specific cancers, thus satisfactory strategies can be developed for the precise treatment of these types of cancer. Currently, there are few targeted drugs to treat gynecologic tumors, and patients with gynecologic cancer often have a poor prognosis because of tumor progression or recurrence. With the application of massively parallel sequencing, a large number of fusion genes have been discovered in gynecologic tumors, and some fusions have been confirmed to be involved in the biological process of tumor progression. To this end, the present article reviews the current research status of all confirmed fusion genes in gynecologic tumors, including their rearrangement mechanism and frequency in ovarian cancer, endometrial cancer, endometrial stromal sarcoma, and other types of uterine tumors. We also describe the mechanisms by which fusion genes are generated and their oncogenic mechanism. Finally, we discuss the prospect of fusion genes as therapeutic targets in gynecologic tumors.
Collapse
Affiliation(s)
- Bingfeng Lu
- Department of Obstetrics and Gynecology, Department of Gynecologic Oncology Research Office, Key Laboratory for Major Obstetric Diseases of Guangdong Province, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Ruqi Jiang
- Department of Obstetrics and Gynecology, Department of Gynecologic Oncology Research Office, Key Laboratory for Major Obstetric Diseases of Guangdong Province, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Bumin Xie
- Department of Obstetrics and Gynecology, Department of Gynecologic Oncology Research Office, Key Laboratory for Major Obstetric Diseases of Guangdong Province, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Wu Wu
- Department of Obstetrics and Gynecology, Department of Gynecologic Oncology Research Office, Key Laboratory for Major Obstetric Diseases of Guangdong Province, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yang Zhao
- Department of Obstetrics and Gynecology, Department of Gynecologic Oncology Research Office, Key Laboratory for Major Obstetric Diseases of Guangdong Province, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
| |
Collapse
|
12
|
Park SJ, Kwon W, Park S, Jeong J, Kim D, Jang S, Kim SY, Sung Y, Kim MO, Choi SK, Ryoo ZY. Jazf1 acts as a regulator of insulin-producing β-cell differentiation in induced pluripotent stem cells and glucose homeostasis in mice. FEBS J 2021; 288:4412-4427. [PMID: 33555104 DOI: 10.1111/febs.15751] [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: 05/18/2020] [Revised: 12/02/2020] [Accepted: 02/04/2021] [Indexed: 12/13/2022]
Abstract
Genetic susceptibility of type 2 diabetes and Juxtaposed with another zinc finger protein 1 (Jazf1) has been reported; however, the precise role of Jazf1 in metabolic processes remains elusive. In this study, using Jazf1-knockout (KO)-induced pluripotent stem cells (iPSC), pancreatic beta cell line MIN6 cells, and Jazf-1 heterozygous KO (Jazf1+/- ) mice, the effect of Jazf1 on gradual differentiation was investigated. We checked the alterations of the genes related with β-cell specification, maturation, and insulin release against glucose treatment by the gain and loss of the Jazf1 gene in the MIN6 cells. Because undifferentiated Jazf1-KO iPSC were not significantly different from wild-type (WT) iPSC, the size and endoderm marker expression after embryoid body (EB) and teratoma formation were investigated. Compared to EB and teratomas formed with WT iPSC, the EB and teratomas from with Jazf1-KO iPSC were smaller, and in teratomas, the expression of proliferation markers was reduced. Moreover, the expression of the gene sets for β-cell differentiation and the levels of insulin and C-peptide secreted by insulin precursor cells were notably reduced in β-cells differentiated from Jazf1-KO iPSC compared with those differentiated from WT iPSC. A comparison of Jazf1+/- and WT mice showed that Jazf1+/- mice had lower levels of serum insulin, pancreatic insulin expression, and decreased pancreatic β-cell size, which resulted in defects in the glucose homeostasis. These findings suggest that Jazf1 plays a pivotal role in the differentiation of β-cells and glucose homeostasis.
Collapse
Affiliation(s)
- Si Jun Park
- School of Life Science, BK21 plus KNU Creative BioResearch Group, Kyungpook National University, Daegu, Korea.,Institute of Life Science and Biotechnology, Kyungpook National University, Daegu, Korea
| | - Wookbong Kwon
- School of Life Science, BK21 plus KNU Creative BioResearch Group, Kyungpook National University, Daegu, Korea.,Division of Biotechnology, DGIST, Daegu, Korea
| | - Song Park
- Core Protein Resources Center, DGIST, Daegu, Korea.,Department of Brain and Cognitive Sciences, DGIST, Daegu, Korea
| | - Jain Jeong
- Core Protein Resources Center, DGIST, Daegu, Korea.,Section of Digestive Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Dongjun Kim
- School of Life Science, BK21 plus KNU Creative BioResearch Group, Kyungpook National University, Daegu, Korea
| | - Soyoung Jang
- School of Life Science, BK21 plus KNU Creative BioResearch Group, Kyungpook National University, Daegu, Korea
| | - Si-Yong Kim
- School of Life Science, BK21 plus KNU Creative BioResearch Group, Kyungpook National University, Daegu, Korea
| | - Yonghun Sung
- Laboratory Animal Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu, Korea
| | - Myoung Ok Kim
- Department of Animal Science and Biotechnology, Kyungpook National University, Sangju, Korea
| | - Seong-Kyoon Choi
- Division of Biotechnology, DGIST, Daegu, Korea.,Core Protein Resources Center, DGIST, Daegu, Korea
| | - Zae Young Ryoo
- School of Life Science, BK21 plus KNU Creative BioResearch Group, Kyungpook National University, Daegu, Korea
| |
Collapse
|
13
|
Wang JG, Jian WJ, Li Y, Zhang J. Nobiletin promotes the pyroptosis of breast cancer via regulation of miR-200b/JAZF1 axis. Kaohsiung J Med Sci 2021; 37:572-582. [PMID: 33728753 DOI: 10.1002/kjm2.12371] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 12/16/2020] [Accepted: 01/14/2021] [Indexed: 12/21/2022] Open
Abstract
Nobiletin is a polymethoxylated flavone present in citrus fruits, which has been reported to have inhibitory effects on tumorigenesis of cancers. However, the biological function of nobiletin in breast cancer (BC) is largely unknown. To investigate the effect of nobiletin on growth of BC cells, the cell viability of BC was measured by MTT assay. In addition, gene and protein expressions were detected by qRT-PCR and western blot, respectively. The apoptosis and pyroptosis of BC cells were tested by flow cytometry. Finally, the correlation between miR-200b and JAZF1 was detected by dual luciferase report. The data indicated that nobiletin inhibited the proliferation of BC cells in a dose-dependent manner. Moreover, miR-200b mimics-induced pyroptosis of BC cells was further increased by nobiletin. Meanwhile, JAZF1 was found to be the target of miR-200b. Moreover, nobiletin induced apoptosis and pyroptosis of BC cells via miR-200b/JAZF1/NF-κB axis. In conclusion, nobiletin inhibited the tumorigenesis of BC via regulation of miR-200b/JAZF1 axis. Thus, nobiletin might serve as a new agent for the treatment of BC.
Collapse
Affiliation(s)
- Ji-Guo Wang
- Department of Oncology, Shenzhen Baoan Hospital of Traditional Chinese Medicine, Shenzhen, China
| | - Wen-Jing Jian
- Department of Breast Surgery, The Second People's Hospital of Shenzhen, Shenzhen, China
| | - Yang Li
- Department of Oncology, Shenzhen Baoan Hospital of Traditional Chinese Medicine, Shenzhen, China
| | - Jing Zhang
- Department of Oncology, Shenzhen Baoan Hospital of Traditional Chinese Medicine, Shenzhen, China
| |
Collapse
|
14
|
JAZF1, A Novel p400/TIP60/NuA4 Complex Member, Regulates H2A.Z Acetylation at Regulatory Regions. Int J Mol Sci 2021; 22:ijms22020678. [PMID: 33445503 PMCID: PMC7826843 DOI: 10.3390/ijms22020678] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 01/07/2021] [Accepted: 01/09/2021] [Indexed: 12/22/2022] Open
Abstract
Histone variants differ in amino acid sequence, expression timing and genomic localization sites from canonical histones and convey unique functions to eukaryotic cells. Their tightly controlled spatial and temporal deposition into specific chromatin regions is accomplished by dedicated chaperone and/or remodeling complexes. While quantitatively identifying the chaperone complexes of many human H2A variants by using mass spectrometry, we also found additional members of the known H2A.Z chaperone complexes p400/TIP60/NuA4 and SRCAP. We discovered JAZF1, a nuclear/nucleolar protein, as a member of a p400 sub-complex containing MBTD1 but excluding ANP32E. Depletion of JAZF1 results in transcriptome changes that affect, among other pathways, ribosome biogenesis. To identify the underlying molecular mechanism contributing to JAZF1's function in gene regulation, we performed genome-wide ChIP-seq analyses. Interestingly, depletion of JAZF1 leads to reduced H2A.Z acetylation levels at > 1000 regulatory sites without affecting H2A.Z nucleosome positioning. Since JAZF1 associates with the histone acetyltransferase TIP60, whose depletion causes a correlated H2A.Z deacetylation of several JAZF1-targeted enhancer regions, we speculate that JAZF1 acts as chromatin modulator by recruiting TIP60's enzymatic activity. Altogether, this study uncovers JAZF1 as a member of a TIP60-containing p400 chaperone complex orchestrating H2A.Z acetylation at regulatory regions controlling the expression of genes, many of which are involved in ribosome biogenesis.
Collapse
|
15
|
Pinto AR, Silva J, Pinto R, Medeiros R. Aggressive prostate cancer phenotype and genome-wide association studies: where are we now? Pharmacogenomics 2020; 21:487-503. [PMID: 32343194 DOI: 10.2217/pgs-2019-0123] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
The majority of prostate cancer (PCa) is indolent, however, a percentage of patients are initially diagnosed with metastatic disease, for which there is a worse prognosis. There is a lack of biomarkers to identify men at greater risk for developing aggressive PCa. Genome-wide association studies (GWAS) scan the genome to search associations of SNPs with specific traits, like cancer. To date, eight GWAS have resulted in the reporting of 16 SNPs associated with aggressive PCa (p < 5.00 × 10-2). Still, validation studies need to be conducted to confirm the obtained results as GWAS can generate false-positive results. Furthermore, post-GWAS studies provide a better understanding of the functional consequences.
Collapse
Affiliation(s)
- Ana R Pinto
- Molecular Oncology & Viral Pathology Group, IPO-Porto Research Center, (CI-IPOP) Portuguese Oncology Institute of Porto (IPO-Porto), Rua Dr. António Bernardino de Almeida, 4200-4072 Porto, Portugal.,ICBAS, Abel Salazar Institute for the Biomedical Sciences, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal
| | - Jani Silva
- Molecular Oncology & Viral Pathology Group, IPO-Porto Research Center, (CI-IPOP) Portuguese Oncology Institute of Porto (IPO-Porto), Rua Dr. António Bernardino de Almeida, 4200-4072 Porto, Portugal
| | - Ricardo Pinto
- Molecular Oncology & Viral Pathology Group, IPO-Porto Research Center, (CI-IPOP) Portuguese Oncology Institute of Porto (IPO-Porto), Rua Dr. António Bernardino de Almeida, 4200-4072 Porto, Portugal
| | - Rui Medeiros
- Molecular Oncology & Viral Pathology Group, IPO-Porto Research Center, (CI-IPOP) Portuguese Oncology Institute of Porto (IPO-Porto), Rua Dr. António Bernardino de Almeida, 4200-4072 Porto, Portugal.,Research Department, Portuguese League Against Cancer (NRNorte), Estrada Interior da Circunvalação, 6657, 4200-172 Porto, Portugal.,CEBIMED, Faculty of Health Sciences, Fernando Pessoa University, Praça 9 de Abril, 349, 4249-004 Porto, Portugal
| |
Collapse
|
16
|
Ehsani R, Drabløs F. Enhanced identification of significant regulators of gene expression. BMC Bioinformatics 2020; 21:134. [PMID: 32252623 PMCID: PMC7132893 DOI: 10.1186/s12859-020-3468-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 03/24/2020] [Indexed: 12/29/2022] Open
Abstract
Background Diseases like cancer will lead to changes in gene expression, and it is relevant to identify key regulatory genes that can be linked directly to these changes. This can be done by computing a Regulatory Impact Factor (RIF) score for relevant regulators. However, this computation is based on estimating correlated patterns of gene expression, often Pearson correlation, and an assumption about a set of specific regulators, normally transcription factors. This study explores alternative measures of correlation, using the Fisher and Sobolev metrics, and an extended set of regulators, including epigenetic regulators and long non-coding RNAs (lncRNAs). Data on prostate cancer have been used to explore the effect of these modifications. Results A tool for computation of RIF scores with alternative correlation measures and extended sets of regulators was developed and tested on gene expression data for prostate cancer. The study showed that the Fisher and Sobolev metrics lead to improved identification of well-documented regulators of gene expression in prostate cancer, and the sets of identified key regulators showed improved overlap with previously defined gene sets of relevance to cancer. The extended set of regulators lead to identification of several interesting candidates for further studies, including lncRNAs. Several key processes were identified as important, including spindle assembly and the epithelial-mesenchymal transition (EMT). Conclusions The study has shown that using alternative metrics of correlation can improve the performance of tools based on correlation of gene expression in genomic data. The Fisher and Sobolev metrics should be considered also in other correlation-based applications.
Collapse
Affiliation(s)
- Rezvan Ehsani
- Department of Mathematics, University of Zabol, Zabol, Iran. .,Department of Bioinformatics, University of Zabol, Zabol, Iran.
| | - Finn Drabløs
- Department of Cancer Research and Molecular Medicine, NTNU - Norwegian University of Science and Technology, NO-7491, Trondheim, Norway.
| |
Collapse
|
17
|
Wu Q, Wu W, Jacevic V, Franca TCC, Wang X, Kuca K. Selective inhibitors for JNK signalling: a potential targeted therapy in cancer. J Enzyme Inhib Med Chem 2020; 35:574-583. [PMID: 31994958 PMCID: PMC7034130 DOI: 10.1080/14756366.2020.1720013] [Citation(s) in RCA: 108] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
c-Jun N-terminal kinase (JNK) signalling regulates both cancer cell apoptosis and survival. Emerging evidence show that JNK promoted tumour progression is involved in various cancers, that include human pancreatic-, lung-, and breast cancer. The pro-survival JNK oncoprotein functions in a cell context- and cell type-specific manner to affect signal pathways that modulate tumour initiation, proliferation, and migration. JNK is therefore considered a potential oncogenic target for cancer therapy. Currently, designing effective and specific JNK inhibitors is an active area in the cancer treatment. Some ATP-competitive inhibitors of JNK, such as SP600125 and AS601245, are widely used in vitro; however, this type of inhibitor lacks specificity as they indiscriminately inhibit phosphorylation of all JNK substrates. Moreover, JNK has at least three isoforms with different functions in cancer development and identifying specific selective inhibitors is crucial for the development of targeted therapy in cancer. Some selective inhibitors of JNK are identified; however, their clinical studies in cancer are relatively less conducted. In this review, we first summarised the function of JNK signalling in cancer progression; there is a focus on the discussion of the novel selective JNK inhibitors as potential targeting therapy in cancer. Finally, we have offered a future perspective of the selective JNK inhibitors in the context of cancer therapies. We hope this review will help to further understand the role of JNK in cancer progression and provide insight into the design of novel selective JNK inhibitors in cancer treatment.
Collapse
Affiliation(s)
- Qinghua Wu
- College of Life Science, Yangtze University, Jingzhou, China.,College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China.,Department of Chemistry, Faculty of Science, University of Hradec Kralove, Hradec Kralove, Czech Republic
| | - Wenda Wu
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China.,Department of Chemistry, Faculty of Science, University of Hradec Kralove, Hradec Kralove, Czech Republic
| | - Vesna Jacevic
- Department of Chemistry, Faculty of Science, University of Hradec Kralove, Hradec Kralove, Czech Republic.,National Poison Control Centre, Military Medical Academy, Belgrade, Serbia.,Medical Faculty of the Military Medical Academy, University of Defence, Belgrade, Serbia
| | - Tanos C C Franca
- Department of Chemistry, Faculty of Science, University of Hradec Kralove, Hradec Kralove, Czech Republic.,Laboratory of Molecular Modeling Applied to the Chemical and Biological Defense, Military Institute of Engineering, Rio de Janeiro, Brazil
| | - Xu Wang
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan, China
| | - Kamil Kuca
- Department of Chemistry, Faculty of Science, University of Hradec Kralove, Hradec Kralove, Czech Republic
| |
Collapse
|
18
|
The role of JNK in prostate cancer progression and therapeutic strategies. Biomed Pharmacother 2020; 121:109679. [DOI: 10.1016/j.biopha.2019.109679] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Revised: 11/10/2019] [Accepted: 11/16/2019] [Indexed: 12/31/2022] Open
|
19
|
Mei JW, Yang ZY, Xiang HG, Bao R, Ye YY, Ren T, Wang XF, Shu YJ. MicroRNA-1275 inhibits cell migration and invasion in gastric cancer by regulating vimentin and E-cadherin via JAZF1. BMC Cancer 2019; 19:740. [PMID: 31357957 PMCID: PMC6664777 DOI: 10.1186/s12885-019-5929-1] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Accepted: 07/12/2019] [Indexed: 12/11/2022] Open
Abstract
Background Emerging evidence has shown that miR-1275 plays a critical role in tumour metastasis and the progression of various types of cancer. In this study, we analysed the role and mechanism of miR-1275 in the progression and prognosis of gastric cancer (GC). Methods Target genes of miR-1275 were identified and verified by luciferase assay and Western blotting. The function of miR-1275 in invasion and metastasis was analysed in vitro and in vivo in nude mice. The signal pathway regulated by miR-1275 was examined by qRT-PCR, Western blotting and chromatin immunoprecipitation analyses. The expression of miR-1275and JAZF1 were measured in specimens of GC and adjacent non cancerous tissues. Results We identified JAZF1 as a direct miR-1275 target. miR-1275 supresses migration and invasion of GC cells in vitro and in vivo, which was restored by JAZF1 overexpression. Moreover, JAZF1 was recognized as a direct regulator of Vimentin. Knocking-down miR-1275 or overexpressing JAZF1 resulted in upregulation of Vimentin but downregulation of E-cadherin. Meanwhile, we validated in 120 GC patients specimens that low miR-1275expression and high JAZF1 mRNA expression levels were closely associated with lymph node metastasis and poor prognosis. The expression of JAZF1 in protein level displayed the correlations with Vimentin but inversely with E-cadherin. Conclusions Increased miR-1275 expression inhibited GC metastasis by regulating vimentin/E-cadherin via direct suppression of JAZF1expression, suggesting that miR-1275 is a tumour-suppressor miRNA with the potential as a prognostic biomarker or therapeutic target in GC. Electronic supplementary material The online version of this article (10.1186/s12885-019-5929-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jia-Wei Mei
- Department of General Surgery and Laboratory of General Surgery, Xinhua Hospital, Affiliated with Shanghai Jiao Tong University, School of Medicine, No. 1665 Kongjiang Road, Shanghai, 200092, China.,Shanghai Key Laboratory of Biliary Tract Disease Research, No. 1665 Kongjiang Road, Shanghai, 200092, China.,Institute of Biliary Tract Disease, Shanghai Jiao Tong University School of Medicine, No. 1665 Kongjiang Road, Shanghai, 200092, China
| | - Zi-Yi Yang
- Department of General Surgery and Laboratory of General Surgery, Xinhua Hospital, Affiliated with Shanghai Jiao Tong University, School of Medicine, No. 1665 Kongjiang Road, Shanghai, 200092, China.,Shanghai Key Laboratory of Biliary Tract Disease Research, No. 1665 Kongjiang Road, Shanghai, 200092, China.,Institute of Biliary Tract Disease, Shanghai Jiao Tong University School of Medicine, No. 1665 Kongjiang Road, Shanghai, 200092, China
| | - Hong-Gang Xiang
- Department of General Surgery, Pudong New Area People's Hospital affiliated to Shanghai University of Medicine and Health Science, No. 490, South Chuanhuan Road, Pudong New Area, Shanghai, 201299, China
| | - Runfa Bao
- Department of General Surgery and Laboratory of General Surgery, Xinhua Hospital, Affiliated with Shanghai Jiao Tong University, School of Medicine, No. 1665 Kongjiang Road, Shanghai, 200092, China.,Shanghai Key Laboratory of Biliary Tract Disease Research, No. 1665 Kongjiang Road, Shanghai, 200092, China.,Institute of Biliary Tract Disease, Shanghai Jiao Tong University School of Medicine, No. 1665 Kongjiang Road, Shanghai, 200092, China
| | - Yuan-Yuan Ye
- Department of General Surgery and Laboratory of General Surgery, Xinhua Hospital, Affiliated with Shanghai Jiao Tong University, School of Medicine, No. 1665 Kongjiang Road, Shanghai, 200092, China.,Shanghai Key Laboratory of Biliary Tract Disease Research, No. 1665 Kongjiang Road, Shanghai, 200092, China.,Institute of Biliary Tract Disease, Shanghai Jiao Tong University School of Medicine, No. 1665 Kongjiang Road, Shanghai, 200092, China
| | - Tai Ren
- Department of General Surgery and Laboratory of General Surgery, Xinhua Hospital, Affiliated with Shanghai Jiao Tong University, School of Medicine, No. 1665 Kongjiang Road, Shanghai, 200092, China.,Shanghai Key Laboratory of Biliary Tract Disease Research, No. 1665 Kongjiang Road, Shanghai, 200092, China.,Institute of Biliary Tract Disease, Shanghai Jiao Tong University School of Medicine, No. 1665 Kongjiang Road, Shanghai, 200092, China
| | - Xue-Feng Wang
- Department of General Surgery and Laboratory of General Surgery, Xinhua Hospital, Affiliated with Shanghai Jiao Tong University, School of Medicine, No. 1665 Kongjiang Road, Shanghai, 200092, China. .,Shanghai Key Laboratory of Biliary Tract Disease Research, No. 1665 Kongjiang Road, Shanghai, 200092, China. .,Institute of Biliary Tract Disease, Shanghai Jiao Tong University School of Medicine, No. 1665 Kongjiang Road, Shanghai, 200092, China.
| | - Yi-Jun Shu
- Department of General Surgery and Laboratory of General Surgery, Xinhua Hospital, Affiliated with Shanghai Jiao Tong University, School of Medicine, No. 1665 Kongjiang Road, Shanghai, 200092, China. .,Shanghai Key Laboratory of Biliary Tract Disease Research, No. 1665 Kongjiang Road, Shanghai, 200092, China. .,Institute of Biliary Tract Disease, Shanghai Jiao Tong University School of Medicine, No. 1665 Kongjiang Road, Shanghai, 200092, China.
| |
Collapse
|