1
|
Islam MSU, Akter N, Zohra FT, Rashid SB, Hasan N, Rahman SM, Sarkar MAR. Genome-wide identification and characterization of cation-proton antiporter (CPA) gene family in rice (Oryza sativa L.) and their expression profiles in response to phytohormones. PLoS One 2025; 20:e0317008. [PMID: 39854520 PMCID: PMC11761165 DOI: 10.1371/journal.pone.0317008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 12/17/2024] [Indexed: 01/26/2025] Open
Abstract
The cation-proton antiporter (CPA) superfamily plays pivotal roles in regulating cellular ion and pH homeostasis in plants. To date, the regulatory functions of CPA family members in rice (Oryza sativa L.) have not been elucidated. In this study, we use rice public data and information techniques, 29 OsCPA candidate genes were identified in the rice japonica variety (referred to as OsCPA) and phylogenetically categorized into K+ efflux antiporter (KEA), Na+/H+ exchanger (NHX), and cation/H+ exchanger (CHX) groups containing 4, 7, and 18 OsCPA genes. The OsCPA proteins were predominantly localized in the plasma membrane and unevenly scattered on 11 chromosomes. The structural analysis of OsCPA proteins revealed higher similarities within groups. Prediction of selection pressure, collinearity, and synteny analysis indicated that all duplicated OsCPA genes had undergone strong purifying selection throughout their evolution. The cis-acting regulatory elements (CAREs) analysis identified 56 CARE motifs responsive to light, tissue, hormones, and stresses. Additionally, 124 miRNA families were identified in the gene promoters, and OsNHX7 was targeted by the highest number of miRNAs (43 miRNAs). Gene Ontology analysis demonstrated the numerous functions of OsCPA genes associated with biological processes (57.14%), cellular components (7.94%), and molecular functions (34.92%). A total of 12 transcription factor families (TFFs), including 40 TFs were identified in gene promoters, with the highest numbers of TFFs (5TFFs) linked to OsCHX13, and OsCHX15. Protein-protein interaction analysis suggested maximum functional similarities between rice and Arabidopsis CPA proteins. Based on expression analysis, OsKEA1, OsKEA2, OsNHX3, and OsNHX7 were frequently expressed in rice tissues. Furthermore, OsNHX3, OsNHX4, OsNHX6, OsNHX7, OsCHX8, and OsCHX17 in abscisic acid, OsKEA1, OsNHX3, and OsCHX8 in gibberellic acid, OsKEA1, OsKEA3, OsNHX1, and OsNHX3 in indole-3-acetic acid treatment were demonstrated as potential candidates in response to hormone. These findings highlight potential candidates for further characterization of OsCPA genes, which may aid in the development of rice varieties.
Collapse
Affiliation(s)
- Md. Shohel Ul Islam
- Laboratory of Functional Genomics and Proteomics, Department of Genetic Engineering and Biotechnology, Faculty of Biological Science and Technology, Jashore University of Science and Technology, Jashore, Bangladesh
| | - Nasrin Akter
- Laboratory of Functional Genomics and Proteomics, Department of Genetic Engineering and Biotechnology, Faculty of Biological Science and Technology, Jashore University of Science and Technology, Jashore, Bangladesh
| | - Fatema Tuz Zohra
- Department of Genetic Engineering and Biotechnology, Faculty of Biological Sciences, University of Rajshahi, Rajshahi, Bangladesh
| | - Shuraya Beente Rashid
- Laboratory of Functional Genomics and Proteomics, Department of Genetic Engineering and Biotechnology, Faculty of Biological Science and Technology, Jashore University of Science and Technology, Jashore, Bangladesh
| | - Naimul Hasan
- Laboratory of Functional Genomics and Proteomics, Department of Genetic Engineering and Biotechnology, Faculty of Biological Science and Technology, Jashore University of Science and Technology, Jashore, Bangladesh
| | - Shaikh Mizanur Rahman
- Laboratory of Functional Genomics and Proteomics, Department of Genetic Engineering and Biotechnology, Faculty of Biological Science and Technology, Jashore University of Science and Technology, Jashore, Bangladesh
| | - Md. Abdur Rauf Sarkar
- Laboratory of Functional Genomics and Proteomics, Department of Genetic Engineering and Biotechnology, Faculty of Biological Science and Technology, Jashore University of Science and Technology, Jashore, Bangladesh
| |
Collapse
|
2
|
Ashkin EL, Tang YJ, Xu H, Hung KL, Belk JA, Cai H, Lopez SS, Dolcen DN, Hebert JD, Li R, Ruiz PA, Keal T, Andrejka L, Chang HY, Petrov DA, Dixon JR, Xu Z, Winslow MM. A STAG2-PAXIP1/PAGR1 axis suppresses lung tumorigenesis. J Exp Med 2025; 222:e20240765. [PMID: 39652422 PMCID: PMC11627241 DOI: 10.1084/jem.20240765] [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: 04/30/2024] [Revised: 09/16/2024] [Accepted: 10/30/2024] [Indexed: 12/12/2024] Open
Abstract
The cohesin complex is a critical regulator of gene expression. STAG2 is the most frequently mutated cohesin subunit across several cancer types and is a key tumor suppressor in lung cancer. Here, we coupled somatic CRISPR-Cas9 genome editing and tumor barcoding with an autochthonous oncogenic KRAS-driven lung cancer model and showed that STAG2 is uniquely tumor-suppressive among all core and auxiliary cohesin components. The heterodimeric complex components PAXIP1 and PAGR1 have highly correlated effects with STAG2 in human lung cancer cell lines, are tumor suppressors in vivo, and are epistatic to STAG2 in oncogenic KRAS-driven lung tumorigenesis in vivo. STAG2 inactivation elicits changes in gene expression, chromatin accessibility, and 3D genome conformation that impact the cancer cell state. Gene expression and chromatin accessibility similarities between STAG2- and PAXIP1-deficient neoplastic cells further relate STAG2-cohesin to PAXIP1/PAGR1. These findings reveal a STAG2-PAXIP1/PAGR1 tumor-suppressive axis and uncover novel PAXIP1-dependent and PAXIP1-independent STAG2-cohesin-mediated mechanisms of lung tumor suppression.
Collapse
Affiliation(s)
- Emily L. Ashkin
- Cancer Biology Program, Stanford University School of Medicine, Stanford, CA, USA
| | - Yuning J. Tang
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Haiqing Xu
- Department of Biology, Stanford University, Stanford, CA, USA
| | - King L. Hung
- Cancer Biology Program, Stanford University School of Medicine, Stanford, CA, USA
- Center for Personal Dynamic Regulomes, Stanford University School of Medicine, Stanford, CA, USA
| | - Julia A. Belk
- Center for Personal Dynamic Regulomes, Stanford University School of Medicine, Stanford, CA, USA
- Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Hongchen Cai
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Steven S. Lopez
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Deniz Nesli Dolcen
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Jess D. Hebert
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Rui Li
- Center for Personal Dynamic Regulomes, Stanford University School of Medicine, Stanford, CA, USA
- Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Paloma A. Ruiz
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Tula Keal
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Laura Andrejka
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Howard Y. Chang
- Center for Personal Dynamic Regulomes, Stanford University School of Medicine, Stanford, CA, USA
- Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Dmitri A. Petrov
- Cancer Biology Program, Stanford University School of Medicine, Stanford, CA, USA
- Department of Biology, Stanford University, Stanford, CA, USA
- Chan Zuckerberg Biohub, San Francisco, CA, USA
| | - Jesse R. Dixon
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Zhichao Xu
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Monte M. Winslow
- Cancer Biology Program, Stanford University School of Medicine, Stanford, CA, USA
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
3
|
Kwon JW, Nguyen TD, Tjota MY, Antic T. GATA3 Expression in Solid Vimentin-Negative Eosinophilic Renal Epithelial Tumors-A Comprehensive Study of 48 Tumors. Int J Surg Pathol 2024:10668969241291885. [PMID: 39523674 DOI: 10.1177/10668969241291885] [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: 11/16/2024]
Abstract
Vimentin-negative solid eosinophilic renal tumors, such as renal oncocytoma, chromophobe renal cell carcinoma (chromophobe RCC), low-grade oncocytic tumor (LOT) of the kidney, and eosinophilic vacuolated tumor (EVT), often present diagnostic challenge to pathologists as they can have significant morphologic overlap. Recent studies have shown that the LOT is consistently positive for GATA3. To test the utility of GATA3 in this potentially challenging diagnostic setting, we investigated GATA3 expression in 48 vimentin-negative solid eosinophilic renal tumors with unequivocal diagnosis, which included 19 LOTs, 3 EVTs, 12 chromophobe RCCs, 11 renal oncocytomas (ROs), and 3 FLCN-mutated renal tumors. GATA3 was at least focally positive in 89% of the LOTs (17/19), 67% of the EVTs (2/3), and 33% of the chromophobe RCCs (4/12) and was negative in all ROs (0/11) and FLCN-mutated renal tumors (0/3). Our study shows that GATA3 can be useful in differentiating between LOT and RO, but it can be focally positive in many tumors of EVT and chromophobe RCC. Therefore, GATA3 could be useful as a panel of immunohistochemical stains that are used in diagnostic algorithm of vimentin-negative solid eosinophilic renal tumors. However, one should not solely rely on GATA3 as vimentin-negative solid eosinophilic renal tumors can have overlapping GATA3 expression.
Collapse
Affiliation(s)
- Jung Woo Kwon
- Department of Pathology, The University of Chicago, Chicago, IL, USA
| | - Tung Duy Nguyen
- Department of Pathology, The University of Chicago, Chicago, IL, USA
| | - Melissa Y Tjota
- Department of Pathology, The University of Chicago, Chicago, IL, USA
| | - Tatjana Antic
- Department of Pathology, The University of Chicago, Chicago, IL, USA
| |
Collapse
|
4
|
Fatema K, Haidar Z, Tanim MTH, Nath SD, Sajib AA. Unveiling the link between arsenic toxicity and diabetes: an in silico exploration into the role of transcription factors. Toxicol Res 2024; 40:653-672. [PMID: 39345741 PMCID: PMC11436564 DOI: 10.1007/s43188-024-00255-y] [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: 10/18/2023] [Revised: 04/10/2024] [Accepted: 07/10/2024] [Indexed: 10/01/2024] Open
Abstract
Arsenic-induced diabetes, despite being a relatively newer finding, is now a growing area of interest, owing to its multifaceted nature of development and the diversity of metabolic conditions that result from it, on top of the already complicated manifestation of arsenic toxicity. Identification and characterization of the common and differentially affected cellular metabolic pathways and their regulatory components among various arsenic and diabetes-associated complications may aid in understanding the core molecular mechanism of arsenic-induced diabetes. This study, therefore, explores the effects of arsenic on human cell lines through 14 transcriptomic datasets containing 160 individual samples using in silico tools to take a systematic, deeper look into the pathways and genes that are being altered. Among these, we especially focused on the role of transcription factors due to their diverse and multifaceted roles in biological processes, aiming to comprehensively investigate the underlying mechanism of arsenic-induced diabetes as well as associated health risks. We present a potential mechanism heavily implying the involvement of the TGF-β/SMAD3 signaling pathway leading to cell cycle alterations and the NF-κB/TNF-α, MAPK, and Ca2+ signaling pathways underlying the pathogenesis of arsenic-induced diabetes. This study also presents novel findings by suggesting potential associations of four transcription factors (NCOA3, PHF20, TFDP1, and TFDP2) with both arsenic toxicity and diabetes; five transcription factors (E2F5, ETS2, EGR1, JDP2, and TFE3) with arsenic toxicity; and one transcription factor (GATA2) with diabetes. The novel association of the transcription factors and proposed mechanism in this study may serve as a take-off point for more experimental evidence needed to understand the in vivo cellular-level diabetogenic effects of arsenic. Supplementary Information The online version contains supplementary material available at 10.1007/s43188-024-00255-y.
Collapse
Affiliation(s)
- Kaniz Fatema
- Department of Genetic Engineering & Biotechnology, University of Dhaka, Dhaka, 1000 Bangladesh
| | - Zinia Haidar
- Department of Genetic Engineering & Biotechnology, University of Dhaka, Dhaka, 1000 Bangladesh
| | - Md Tamzid Hossain Tanim
- Department of Genetic Engineering & Biotechnology, University of Dhaka, Dhaka, 1000 Bangladesh
| | - Sudipta Deb Nath
- Department of Genetic Engineering & Biotechnology, University of Dhaka, Dhaka, 1000 Bangladesh
| | - Abu Ashfaqur Sajib
- Department of Genetic Engineering & Biotechnology, University of Dhaka, Dhaka, 1000 Bangladesh
| |
Collapse
|
5
|
Opris CE, Suciu H, Flamand S, Opris CI, Hamida AH, Gurzu S. Update on the genetic profile of mitral valve development and prolapse. Pathol Res Pract 2024; 262:155535. [PMID: 39182449 DOI: 10.1016/j.prp.2024.155535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 07/21/2024] [Accepted: 08/11/2024] [Indexed: 08/27/2024]
Abstract
The purpose of this review is to present a comprehensive overview of the literature published up to February 2024 on the PubMed database regarding the development of mitral valve disease, with detailed reference to mitral valve prolapse, from embryology to a genetic profile. Out of the 3291 publications that deal with mitral valve embryology, 215 refer to mitral valve genetics and 83 were selected for further analysis. After reviewing these data, we advocate for the importance of a gene-based therapy that should be available soon, to prevent or treat non-invasively the valvular degeneration.
Collapse
Affiliation(s)
- Carmen Elena Opris
- Department of Adult and Children Cardiovascular Recovery, Emergency Institute for Cardio-Vascular Diseases and Transplantation, Targu Mures 540139, Romania; Department of Pathology, George Emil Palade University of Medicine, Pharmacy, Science and Technology, Targu Mures , Romania; Department of Cardiovascular Surgery, Emergency University Hospital, Romania
| | - Horatiu Suciu
- Department of Surgery, George Emil Palade University of Medicine, Pharmacy, Sciences and Technology, Targu Mures 540139, Romania; Romanian Academy of Medical Sciences, Romania; Department of Cardiovascular Surgery, Emergency University Hospital, Romania
| | - Sanziana Flamand
- Department of Surgery, George Emil Palade University of Medicine, Pharmacy, Sciences and Technology, Targu Mures 540139, Romania; Department of Cardiovascular Surgery, Emergency University Hospital, Romania
| | - Cosmin Ioan Opris
- Department of Surgery, George Emil Palade University of Medicine, Pharmacy, Sciences and Technology, Targu Mures 540139, Romania; Department of Cardiovascular Surgery, Emergency University Hospital, Romania
| | - Al Hussein Hamida
- Department of Surgery, George Emil Palade University of Medicine, Pharmacy, Sciences and Technology, Targu Mures 540139, Romania
| | - Simona Gurzu
- Department of Pathology, George Emil Palade University of Medicine, Pharmacy, Science and Technology, Targu Mures , Romania; Romanian Academy of Medical Sciences, Romania; Research Center for Oncopathology and Translational Medicine (CCOMT), George Emil Palade University of Medicine, Pharmacy, Science and Technology, Targu Mures, Romania.
| |
Collapse
|
6
|
Peretz CAC, Kennedy VE, Walia A, Delley CL, Koh A, Tran E, Clark IC, Hayford CE, D'Amato C, Xue Y, Fontanez KM, May-Zhang AA, Smithers T, Agam Y, Wang Q, Dai HP, Roy R, Logan AC, Perl AE, Abate A, Olshen A, Smith CC. Multiomic single cell sequencing identifies stemlike nature of mixed phenotype acute leukemia. Nat Commun 2024; 15:8191. [PMID: 39294124 PMCID: PMC11411136 DOI: 10.1038/s41467-024-52317-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 08/30/2024] [Indexed: 09/20/2024] Open
Abstract
Despite recent work linking mixed phenotype acute leukemia (MPAL) to certain genetic lesions, specific driver mutations remain undefined for a significant proportion of patients and no genetic subtype is predictive of clinical outcomes. Moreover, therapeutic strategy for MPAL remains unclear, and prognosis is overall poor. We performed multiomic single cell profiling of 14 newly diagnosed adult MPAL patients to characterize the inter- and intra-tumoral transcriptional, immunophenotypic, and genetic landscapes of MPAL. We show that neither genetic profile nor transcriptome reliably correlate with specific MPAL immunophenotypes. Despite this, we find that MPAL blasts express a shared stem cell-like transcriptional profile indicative of high differentiation potential. Patients with the highest differentiation potential demonstrate inferior survival in our dataset. A gene set score, MPAL95, derived from genes highly enriched in the most stem-like MPAL cells, is applicable to bulk RNA sequencing data and is predictive of survival in an independent patient cohort, suggesting a potential strategy for clinical risk stratification.
Collapse
Affiliation(s)
- Cheryl A C Peretz
- Division of Hematology and Oncology, Department of Pediatrics, University of California San Francisco, San Francisco, CA, USA
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA
| | - Vanessa E Kennedy
- Division of Hematology and Oncology, Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Anushka Walia
- Division of Hematology and Oncology, Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Cyrille L Delley
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA, USA
| | - Andrew Koh
- Division of Hematology and Oncology, Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Elaine Tran
- Division of Hematology and Oncology, Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Iain C Clark
- Department of Bioengineering, University of California Berkeley, Berkeley, CA, USA
| | | | | | - Yi Xue
- Fluent Biosciences Inc., Watertown, MA, USA
| | | | | | | | - Yigal Agam
- Fluent Biosciences Inc., Watertown, MA, USA
| | - Qian Wang
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, People's Republic of China
| | - Hai-Ping Dai
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, People's Republic of China
| | - Ritu Roy
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA
| | - Aaron C Logan
- Division of Hematology and Oncology, Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Alexander E Perl
- Department of Medicine, Division of Hematology-Oncology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Adam Abate
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA, USA
| | - Adam Olshen
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, CA, USA
| | - Catherine C Smith
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA.
- Division of Hematology and Oncology, Department of Medicine, University of California San Francisco, San Francisco, CA, USA.
| |
Collapse
|
7
|
Yu W, Lin X, Leng S, Hou Y, Dang Z, Xue S, Li N, Zhang F. The PRC2 complex epigenetically silences GATA4 to suppress cellular senescence and promote the progression of breast cancer. Transl Oncol 2024; 46:102014. [PMID: 38843657 PMCID: PMC11214403 DOI: 10.1016/j.tranon.2024.102014] [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: 04/03/2024] [Revised: 05/17/2024] [Accepted: 05/27/2024] [Indexed: 06/19/2024] Open
Abstract
BACKGROUND The transcription factor GATA4 is pivotal in cancer development but is often silenced through mechanisms like DNA methylation and histone modifications. This silencing suppresses the transcriptional activity of GATA4, disrupting its normal functions and promoting cancer progression. However, the precise molecular mechanisms and implications of GATA4 silencing in tumorigenesis remain unclear. Here, we aim to elucidate the mechanisms underlying GATA4 silencing and explore its role in breast cancer progression and its potential as a therapeutic target. METHODS The GATA4-breast cancer prognosis link was explored via bioinformatics analyses, with GATA4 expression measured in breast tissues. Functional gain/loss experiments were performed to gauge GATA4's impact on breast cancer cell malignancy. GATA4-PRC2 complex interaction was analyzed using silver staining and mass spectrometry. Chromatin immunoprecipitation, coupled with high-throughput sequencing, was used to identify GATA4-regulated downstream target genes. The in vitro findings were validated in an in situ breast cancer xenograft mouse model. RESULTS GATA4 mutation and different breast cancer subtypes were correlated, suggesting its involvement in disease progression. GATA4 suppressed cell proliferation, invasion, and migration while inducing apoptosis and senescence in breast cancer cells. The GATA4-PRC2 complex interaction silenced GATA4 expression, which altered the regulation of FAS, a GATA4 downstream gene. In vivo experiments verified that GATA4 inhibits tumor growth, suggesting its regulatory function in tumorigenesis. CONCLUSIONS This comprehensive study highlights the epigenetic regulation of GATA4 and its impact on breast cancer development, highlighting the PRC2-GATA4-FAS pathway as a potential target for therapeutic interventions in breast cancers.
Collapse
Affiliation(s)
- Wenqian Yu
- Research Center of Translational Medicine, Department of Cardiac Surgery, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250013, China
| | - Xiaona Lin
- Department of Cardiovascular Surgery, Shandong Second Provincial General Hospital, Jinan, Shandong 250022, China
| | - Shuai Leng
- Research Center of Translational Medicine, Department of Cardiac Surgery, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250013, China
| | - Yiming Hou
- Research Center of Translational Medicine, Department of Cardiac Surgery, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250013, China
| | - Zhiqiao Dang
- Research Center of Translational Medicine, Department of Cardiac Surgery, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250013, China
| | - Shishan Xue
- Research Center of Translational Medicine, Department of Cardiac Surgery, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250013, China
| | - Na Li
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Shandong University, No. 4, Duanxin West Road, Jinan, Shandong 250022, China; Center of Clinical Laboratory, Shandong Second Provincial General Hospital, Jinan, Shandong 250022, China.
| | - Fengquan Zhang
- Research Center of Translational Medicine, Department of Cardiac Surgery, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250013, China.
| |
Collapse
|
8
|
Yoo KC, Kim DH, Park S, Yun H, Ryu DH, Lee J, Son SM. Gastric Metastasis Mimicking Early Gastric Cancer from Invasive Ductal Carcinoma of the Breast: Case Report and Literature Review. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:980. [PMID: 38929597 PMCID: PMC11205986 DOI: 10.3390/medicina60060980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 05/28/2024] [Accepted: 06/10/2024] [Indexed: 06/28/2024]
Abstract
Backgound and Objectives: Gastric metastasis from invasive ductal breast cancer (BC) is rare. It mainly occurs in patients with lobular BC. The occurrence of multiple metastases is typically observed several years after the primary diagnosis. Endoscopic findings of gastric metastasis of the BC were usually the linitis plastic type. Case presentation: A 72-year-old women who underwent right modified radical mastectomy (MRM) 10 month ago was referred after being diagnosed with early gastric cancer (EGC) during systemic chemotherapy. EGC type I was found at gastric fundus, and pathologic finding showed poorly differentiated adenocarcinoma. Metachronous double primary tumor EGC was considered. Management and Outcome: A laparoscopic total gastrectomy was performed, and postoperative pathology revealed submucosa invasion and two lymph node metastases. A pathologic review that focused on immunohistochemical studies of selected antibodies such as GATA binding protein 3 (GATA3), gross cystic disease fluid protein-15 (GCDFP-15), cytokeratin 7 (CK7) was performed again, comparing previous results. As a result, gastric metastasis from BC was diagnosed. After totally laparoscopic total gastrectomy, palliative first-line chemotherapy with paclitaxel/CDDP was performed. Two months after gastrectomy, she was diagnosed with para-aortic lymph node metastasis and multiple bone metastases. She expired six months after gastrectomy. Conclusions: Gastric metastasis from invasive ductal carcinoma of the breast, which is clinically manifested as EGC, is a very rare condition. If there is a history of BC, careful pathological review will be required.
Collapse
Affiliation(s)
- Kwon Cheol Yoo
- Department of Surgery, Chungbuk National University Hospital, Cheongju 28644, Republic of Korea; (K.C.Y.)
- Department of Surgery, Chungbuk National University College of Medicine, Cheongju 28644, Republic of Korea
| | - Dae Hoon Kim
- Department of Surgery, Chungbuk National University Hospital, Cheongju 28644, Republic of Korea; (K.C.Y.)
- Department of Surgery, Chungbuk National University College of Medicine, Cheongju 28644, Republic of Korea
| | - Sungmin Park
- Department of Surgery, Chungbuk National University Hospital, Cheongju 28644, Republic of Korea; (K.C.Y.)
- Department of Surgery, Chungbuk National University College of Medicine, Cheongju 28644, Republic of Korea
| | - HyoYung Yun
- Department of Surgery, Chungbuk National University Hospital, Cheongju 28644, Republic of Korea; (K.C.Y.)
- Department of Surgery, Chungbuk National University College of Medicine, Cheongju 28644, Republic of Korea
| | - Dong Hee Ryu
- Department of Surgery, Chungbuk National University Hospital, Cheongju 28644, Republic of Korea; (K.C.Y.)
- Department of Surgery, Chungbuk National University College of Medicine, Cheongju 28644, Republic of Korea
| | - Jisun Lee
- Department of Radiology, Chungbuk National University Hospital, Cheongju 28644, Republic of Korea
- Department of Radiology, Chungbuk National University College of Medicine, Cheongju 28644, Republic of Korea
| | - Seung-Myoung Son
- Department of Pathology, Chungbuk National University Hospital, Cheongju 28644, Republic of Korea
- Department of Pathology, Chungbuk National University College of Medicine, Cheongju 28644, Republic of Korea
| |
Collapse
|
9
|
The role of GATA family transcriptional factors in haematological malignancies: A review: Retraction. Medicine (Baltimore) 2024; 103:e38232. [PMID: 38701322 PMCID: PMC11062697 DOI: 10.1097/md.0000000000038232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/05/2024] Open
|
10
|
O'Brien G, Cecotka A, Manola KN, Pagoni MN, Polanska J, Badie C. Epigenetic signature of ionizing radiation in therapy-related AML patients. Heliyon 2024; 10:e23244. [PMID: 38163095 PMCID: PMC10757008 DOI: 10.1016/j.heliyon.2023.e23244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 10/26/2023] [Accepted: 11/29/2023] [Indexed: 01/03/2024] Open
Abstract
Therapy-related acute myeloid leukaemia (t-AML) is a late side effect of previous chemotherapy (ct-AML) and/or radiotherapy (rt-AML) or immunosuppressive treatment. t-AMLs, which account for ∼10-20 % of all AML cases, are extremely aggressive and have a poor prognosis compared to de novo AML. Our hypothesis is that exposure to radiation causes genome-wide epigenetic changes in rt-AML. An epigenome-wide association study was undertaken, measuring over 850K methylation sites across the genome from fifteen donors (five healthy, five de novo, and five t-AMLs). The study predominantly focussed on 94K sites that lie in CpG-rich gene promoter regions. Genome-wide hypomethylation was discovered in AML, primarily in intergenic regions. Additionally, genes specific to AML were identified with promoter hypermethylation. A two-step validation was conducted, both internally, using pyrosequencing to measure methylation levels in specific regions across fifteen primary samples, and externally, with an additional eight AML samples. We demonstrated that the MEST and GATA5 gene promoters, which were previously identified as tumour suppressors, were noticeably hypermethylated in rt-AML, as opposed to other subtypes of AML and control samples. These may indicate the epigenetic involvement in the development of rt-AML at the molecular level and could serve as potential targets for drug therapy in rt-AML.
Collapse
Affiliation(s)
- Gráinne O'Brien
- Cancer Mechanisms and Biomarkers Group, Radiation Effects Department Radiation, Chemical & Environmental Hazards, Harwell Campus, Chilton, Didcot, Oxfordshire OX11 ORQ, UK Health Security Agency (UKHSA), United Kingdom
| | - Agnieszka Cecotka
- Department of Data Science and Engineering, Silesian University of Technology, 44-121 Gliwice, Poland
| | - Kalliopi N. Manola
- Department of Biodiagnostic Sciences and Technologies, INRASTES, National Centre for Research' Demokritos', 15341 Agia Paraskevi, Greece
| | - Maria N. Pagoni
- Hematology-Lymphomas Department - BMT Unit, Evangelismos Hospital, 10676 Athens, Greece
| | - Joanna Polanska
- Department of Data Science and Engineering, Silesian University of Technology, 44-121 Gliwice, Poland
| | - Christophe Badie
- Cancer Mechanisms and Biomarkers Group, Radiation Effects Department Radiation, Chemical & Environmental Hazards, Harwell Campus, Chilton, Didcot, Oxfordshire OX11 ORQ, UK Health Security Agency (UKHSA), United Kingdom
- Environmental Research Group Within the School of Public Health, Faculty of Medicine at Imperial College of Science, Technology and Medicine, London W12 0BZ, United Kingdom
| |
Collapse
|
11
|
Saatci O, Sahin O. TACC3: a multi-functional protein promoting cancer cell survival and aggressiveness. Cell Cycle 2023; 22:2637-2655. [PMID: 38197196 PMCID: PMC10936615 DOI: 10.1080/15384101.2024.2302243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 12/30/2023] [Accepted: 01/02/2024] [Indexed: 01/11/2024] Open
Abstract
TACC3 is the most oncogenic member of the transforming acidic coiled-coil domain-containing protein (TACC) family. It is one of the major recruitment factors of distinct multi-protein complexes. TACC3 is localized to spindles, centrosomes, and nucleus, and regulates key oncogenic processes, including cell proliferation, migration, invasion, and stemness. Recently, TACC3 inhibition has been identified as a vulnerability in highly aggressive cancers, such as cancers with centrosome amplification (CA). TACC3 has spatiotemporal functions throughout the cell cycle; therefore, targeting TACC3 causes cell death in mitosis and interphase in cancer cells with CA. In the clinics, TACC3 is highly expressed and associated with worse survival in multiple cancers. Furthermore, TACC3 is a part of one of the most common fusions of FGFR, FGFR3-TACC3 and is important for the oncogenicity of the fusion. A detailed understanding of the regulation of TACC3 expression, its key partners, and molecular functions in cancer cells is vital for uncovering the most vulnerable tumors and maximizing the therapeutic potential of targeting this highly oncogenic protein. In this review, we summarize the established and emerging interactors and spatiotemporal functions of TACC3 in cancer cells, discuss the potential of TACC3 as a biomarker in cancer, and therapeutic potential of its inhibition.
Collapse
Affiliation(s)
- Ozge Saatci
- Department of Biochemistry and Molecular Biology, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, USA
| | - Ozgur Sahin
- Department of Biochemistry and Molecular Biology, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, USA
| |
Collapse
|
12
|
Palumbo GA, Duminuco A. Myelofibrosis: In Search for BETter Targeted Therapies. J Clin Oncol 2023; 41:5044-5048. [PMID: 37751563 DOI: 10.1200/jco.23.00833] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Revised: 07/22/2023] [Accepted: 07/31/2023] [Indexed: 09/28/2023] Open
Affiliation(s)
- Giuseppe A Palumbo
- Department of Scienze Mediche, Chirurgiche e Tecnologie Avanzate "G.F. Ingrassia," University of Catania, Catania, Italy
| | - Andrea Duminuco
- Postgraduate School of Hematology, University of Catania, Catania, Italy
- Department of Haematology, Guy's and St Thomas' NHS Foundation Trust, London, United Kingdom
| |
Collapse
|
13
|
Martin-Hernandez R, Espeso-Gil S, Domingo C, Latorre P, Hervas S, Hernandez Mora JR, Kotelnikova E. Machine learning combining multi-omics data and network algorithms identifies adrenocortical carcinoma prognostic biomarkers. Front Mol Biosci 2023; 10:1258902. [PMID: 38028548 PMCID: PMC10658191 DOI: 10.3389/fmolb.2023.1258902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 10/06/2023] [Indexed: 12/01/2023] Open
Abstract
Background: Rare endocrine cancers such as Adrenocortical Carcinoma (ACC) present a serious diagnostic and prognostication challenge. The knowledge about ACC pathogenesis is incomplete, and patients have limited therapeutic options. Identification of molecular drivers and effective biomarkers is required for timely diagnosis of the disease and stratify patients to offer the most beneficial treatments. In this study we demonstrate how machine learning methods integrating multi-omics data, in combination with system biology tools, can contribute to the identification of new prognostic biomarkers for ACC. Methods: ACC gene expression and DNA methylation datasets were downloaded from the Xena Browser (GDC TCGA Adrenocortical Carcinoma cohort). A highly correlated multi-omics signature discriminating groups of samples was identified with the data integration analysis for biomarker discovery using latent components (DIABLO) method. Additional regulators of the identified signature were discovered using Clarivate CBDD (Computational Biology for Drug Discovery) network propagation and hidden nodes algorithms on a curated network of molecular interactions (MetaBase™). The discriminative power of the multi-omics signature and their regulators was delineated by training a random forest classifier using 55 samples, by employing a 10-fold cross validation with five iterations. The prognostic value of the identified biomarkers was further assessed on an external ACC dataset obtained from GEO (GSE49280) using the Kaplan-Meier estimator method. An optimal prognostic signature was finally derived using the stepwise Akaike Information Criterion (AIC) that allowed categorization of samples into high and low-risk groups. Results: A multi-omics signature including genes, micro RNA's and methylation sites was generated. Systems biology tools identified additional genes regulating the features included in the multi-omics signature. RNA-seq, miRNA-seq and DNA methylation sets of features revealed a high power to classify patients from stages I-II and stages III-IV, outperforming previously identified prognostic biomarkers. Using an independent dataset, associations of the genes included in the signature with Overall Survival (OS) data demonstrated that patients with differential expression levels of 8 genes and 4 micro RNA's showed a statistically significant decrease in OS. We also found an independent prognostic signature for ACC with potential use in clinical practice, combining 9-gene/micro RNA features, that successfully predicted high-risk ACC cancer patients. Conclusion: Machine learning and integrative analysis of multi-omics data, in combination with Clarivate CBDD systems biology tools, identified a set of biomarkers with high prognostic value for ACC disease. Multi-omics data is a promising resource for the identification of drivers and new prognostic biomarkers in rare diseases that could be used in clinical practice.
Collapse
|
14
|
Pihlajoki M, Eloranta K, Nousiainen R, Väyrynen V, Soini T, Kyrönlahti A, Parkkila S, Kanerva J, Wilson DB, Pakarinen MP, Heikinheimo M. Biology of childhood hepatoblastoma and the search for novel treatments. Adv Biol Regul 2023; 91:100997. [PMID: 39492287 DOI: 10.1016/j.jbior.2023.100997] [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: 10/09/2023] [Accepted: 10/17/2023] [Indexed: 11/05/2024]
Abstract
Our research laboratory has a longstanding interest in developmental disorders and embryonic tumors, and recent efforts have focused on the pathogenesis of pediatric liver tumors. This review focuses on hepatoblastoma (HB), the most common pediatric liver malignancy. Despite advances in treatment, patients with metastatic HB have a poor prognosis, and survivors often have permanent side effects attributable to chemotherapy. In an effort to improve survival and lessen long-term complications of HB, we have searched for novel molecular vulnerabilities using a combination of patient derived cell lines, metabolomics, and RNA sequencing of human samples at diagnosis and follow-up. These studies have shed light on pathogenesis and identified putative targets for future therapies in children with advanced HB.
Collapse
Affiliation(s)
- Marjut Pihlajoki
- Pediatric Research Center, Children's Hospital, University of Helsinki and Helsinki University Hospital, Helsinki, Finland.
| | - Katja Eloranta
- Pediatric Research Center, Children's Hospital, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Ruth Nousiainen
- Pediatric Research Center, Children's Hospital, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Ville Väyrynen
- Pediatric Research Center, Children's Hospital, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Tea Soini
- Pediatric Research Center, Children's Hospital, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Antti Kyrönlahti
- Pediatric Research Center, Children's Hospital, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Seppo Parkkila
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland; FICAN Mid, Tampere University, Tampere, Finland; Fimlab Ltd, Tampere University Hospital, Tampere, Finland
| | - Jukka Kanerva
- Pediatric Research Center, Children's Hospital, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - David B Wilson
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, United States; Department of Pediatrics, Washington University in St. Louis, St. Louis, United States
| | - Mikko P Pakarinen
- Pediatric Research Center, Children's Hospital, University of Helsinki and Helsinki University Hospital, Helsinki, Finland; Section of Pediatric Surgery, Pediatric Liver and Gut Research Group, Pediatric Research Department of Women's Health, Karolinska Institute, Stockholm, Sweden
| | - Markku Heikinheimo
- Pediatric Research Center, Children's Hospital, University of Helsinki and Helsinki University Hospital, Helsinki, Finland; Department of Pediatrics, Washington University in St. Louis, St. Louis, United States; Faculty of Medicine and Health Technology, Center for Child, Adolescent, and Maternal Health Research, Tampere University, Tampere, Finland
| |
Collapse
|
15
|
Williams RTP, King DC, Mastroianni IR, Hill JL, Apenes NW, Ramirez G, Miner EC, Moore A, Coleman K, Nishimura EO. Transcriptome profiling of the Caenorhabditis elegans intestine reveals that ELT-2 negatively and positively regulates intestinal gene expression within the context of a gene regulatory network. Genetics 2023; 224:iyad088. [PMID: 37183501 PMCID: PMC10411582 DOI: 10.1093/genetics/iyad088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 04/28/2023] [Accepted: 04/30/2023] [Indexed: 05/16/2023] Open
Abstract
ELT-2 is the major transcription factor (TF) required for Caenorhabditis elegans intestinal development. ELT-2 expression initiates in embryos to promote development and then persists after hatching through the larval and adult stages. Though the sites of ELT-2 binding are characterized and the transcriptional changes that result from ELT-2 depletion are known, an intestine-specific transcriptome profile spanning developmental time has been missing. We generated this dataset by performing Fluorescence Activated Cell Sorting on intestine cells at distinct developmental stages. We analyzed this dataset in conjunction with previously conducted ELT-2 studies to evaluate the role of ELT-2 in directing the intestinal gene regulatory network through development. We found that only 33% of intestine-enriched genes in the embryo were direct targets of ELT-2 but that number increased to 75% by the L3 stage. This suggests additional TFs promote intestinal transcription especially in the embryo. Furthermore, only half of ELT-2's direct target genes were dependent on ELT-2 for their proper expression levels, and an equal proportion of those responded to elt-2 depletion with over-expression as with under-expression. That is, ELT-2 can either activate or repress direct target genes. Additionally, we observed that ELT-2 repressed its own promoter, implicating new models for its autoregulation. Together, our results illustrate that ELT-2 impacts roughly 20-50% of intestine-specific genes, that ELT-2 both positively and negatively controls its direct targets, and that the current model of the intestinal regulatory network is incomplete as the factors responsible for directing the expression of many intestinal genes remain unknown.
Collapse
Affiliation(s)
- Robert T P Williams
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO 80523, USA
| | - David C King
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO 80523, USA
| | - Izabella R Mastroianni
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO 80523, USA
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jessica L Hill
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO 80523, USA
| | - Nicolai W Apenes
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO 80523, USA
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Gabriela Ramirez
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO 80523, USA
- Department of Cell and Molecular Biology, Colorado State University, Fort Collins, CO 80523, USA
| | - E Catherine Miner
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO 80523, USA
- College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO 80523, USA
| | - Andrew Moore
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO 80523, USA
| | - Karissa Coleman
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO 80523, USA
| | - Erin Osborne Nishimura
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO 80523, USA
| |
Collapse
|
16
|
Baban F, Koepplin JW, Ahmad M, Clarke-Brodber AL, Bois MC, Hartley CP, Sturgis CD. TRPS1 outperforms GATA3 in pleural effusions with metastatic breast carcinoma versus mesothelioma. Diagn Cytopathol 2023. [PMID: 37096814 DOI: 10.1002/dc.25148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 03/28/2023] [Accepted: 04/17/2023] [Indexed: 04/26/2023]
Abstract
INTRODUCTION In evaluating malignant pleural fluid cytology, metastatic adenocarcinomas and mesotheliomas are often differential diagnoses. GATA binding protein 3 (GATA3) has historically been used to confirm metastatic breast carcinomas; however, GATA3 has low specificity if mesothelioma is included in differential diagnoses. Trichorhinophalangeal syndrome type 1 (TRPS1) protein is expressed in all types of breast carcinomas, with reported high specificity and sensitivity. We investigated the performance of TRPS1 immunohistochemistry (IHC) and compared it to GATA3 in pleural fluids diagnosed with metastatic breast carcinoma and mesothelioma. METHODS Thirty-six consecutive ThinPrep pleural fluids and 4 pleural fine needle aspirations (FNAs) with diagnoses of metastatic breast carcinoma (21) and mesothelioma (19) were retrieved, and IHC with TRPS1 and GATA3 was performed on all. Immunoreactivity scores for TRPS1 were calculated by multiplying percentage of immunoreactive cells by staining intensity. Immunoreactivity scores were negative if 0 or 1, low positive if 2, intermediate positive if 3 or 4, or high positive if 6 or 9. Nuclear immunoreactivity of ≥10% with at least moderate intensity was judged GATA3 positive. RESULTS GATA3 showed immunoreactivity in all metastatic breast carcinomas and 84% of mesotheliomas. TRPS1 was immunoreactive in all breast carcinoma cases (18 with a score of 9 and 3 with a score of 6). TRPS1 showed low positivity in 5% of mesothelioma cases with all other cases being negative. CONCLUSION When cytomorphologic differential diagnoses of mesothelioma exist, TRPS1 is a more specific marker than GATA3 for confirmation of metastatic breast carcinoma in pleural fluid cytology.
Collapse
Affiliation(s)
- Farah Baban
- Department of Laboratory Medicine and Pathology, Division of Anatomic Pathology-Mayo Clinic, Rochester, Minnesota, USA
| | - Justin W Koepplin
- Department of Laboratory Medicine and Pathology, Division of Anatomic Pathology-Mayo Clinic, Rochester, Minnesota, USA
| | - Muhammad Ahmad
- Department of Laboratory Medicine and Pathology, Division of Anatomic Pathology-Mayo Clinic, Rochester, Minnesota, USA
| | - Anna-Lee Clarke-Brodber
- Department of Laboratory Medicine and Pathology, Division of Anatomic Pathology-Mayo Clinic, Rochester, Minnesota, USA
| | - Melanie C Bois
- Department of Laboratory Medicine and Pathology, Division of Anatomic Pathology-Mayo Clinic, Rochester, Minnesota, USA
| | - Christopher P Hartley
- Department of Laboratory Medicine and Pathology, Division of Anatomic Pathology-Mayo Clinic, Rochester, Minnesota, USA
| | - Charles D Sturgis
- Department of Laboratory Medicine and Pathology, Division of Anatomic Pathology-Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
17
|
Pérez-Montiel D, Olivares-Mundo A, Jiménez-Ríos MA, Morera CS, Scavuzzo A, Orozco-Mora A, Prada D, Cantú-de León D. Influence of luminal and basal subtype in prognosis of high-grade non muscle invasive urothelial carcinoma. Ann Diagn Pathol 2023; 63:152081. [PMID: 36680930 PMCID: PMC11210443 DOI: 10.1016/j.anndiagpath.2022.152081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Accepted: 12/19/2022] [Indexed: 01/19/2023]
Abstract
BACKGROUND Recent studies have shown that the classification of high-grade urothelial carcinoma non-muscle invasive (HGBCNMI) based on molecular subtypes might be a valuable strategy to identify patients with a worse clinical prognosis. OBJECTIVE Determine the effect of the luminal and basal molecular subtype determined by immunistochemical on prognosis in patients with HGBC in Mexican population. METHODS Phenotypes were evaluated by immunohistochemical staining of luminal (GATA3, FOXA1) and basal (CK5/6, CK14) markers in paraffin-embedded tissue samples from 45 patients with a diagnosis of HGBCNMI treated at Instituto Nacional de Cancerología-México (INCan) between 2009 and 2019. The association with prognosis was evaluated using Kaplan-Meier curves and multivariable-adjusted Cox models. RESULTS HGBCNMI patients showed mean age of 58.77 years (SD: ±12.08 years). We identified expression of the luminal molecular subtype in 35 cases (77.78 %), and 10 cases (22.22 %) with "combined" expression of the molecular subtype (basal and luminal expression). The combined phenotype was statistically more frequent in metastatic cases (p-value = 0.028). In Kaplan-Meier curves, combined expression of luminal and basal molecular markers was associated with disease progression (p-value = 0.002, log-rank test). Cox regression models confirmed this association, which was not influenced by age (p-value = 0.007) or gender (p-value = 0.007). No association of phenotypes with overall survival (p-value = 0.860) or relapse (p-value = 0.5) was observed. CONCLUSION The combined expression of immunohistochemical markers of the luminal and basal subtype might be considered as predictor for disease progression in patients with HGBCNMI in Mexican population.
Collapse
Affiliation(s)
- Delia Pérez-Montiel
- Departamento de Patología, Instituto Nacional de Cancerología, San Fernando 22, Colonia Sección XVI, Tlalpan, Ciudad de México 14050, Mexico
| | - Anny Olivares-Mundo
- Subdirección de Investigación Básica, Instituto Nacional de Cancerología, San Fernando 22, Colonia Sección XVI, Tlalpan, Ciudad de México 14050, Mexico
| | - Miguel A Jiménez-Ríos
- Departamento de Urología, Instituto Nacional de Cancerología, San Fernando 22, Colonia Sección XVI, Tlalpan, Ciudad de México 14050, Mexico
| | - Carolina Silva Morera
- Departamento de Patología, Instituto Nacional de Cancerología, San Fernando 22, Colonia Sección XVI, Tlalpan, Ciudad de México 14050, Mexico
| | - Anna Scavuzzo
- Departamento de Urología, Instituto Nacional de Cancerología, San Fernando 22, Colonia Sección XVI, Tlalpan, Ciudad de México 14050, Mexico
| | - Alicia Orozco-Mora
- Subdirección de Investigación Básica, Instituto Nacional de Cancerología, San Fernando 22, Colonia Sección XVI, Tlalpan, Ciudad de México 14050, Mexico
| | - Diddier Prada
- Subdirección de Investigación Básica, Instituto Nacional de Cancerología, San Fernando 22, Colonia Sección XVI, Tlalpan, Ciudad de México 14050, Mexico; Department of Environmental Health Sciences, Columbia University Mailman School of Public Health, New York, 10032, USA.
| | - David Cantú-de León
- Dirección de Investigación, Instituto Nacional de Cancerología, San Fernando 22, Colonia Sección XVI, Tlalpan, Ciudad de México 14050, Mexico.
| |
Collapse
|
18
|
Carpio-Escalona LV, González-de-Olano D. Immunological and Non-Immunological Risk Factors in Anaphylaxis. CURRENT TREATMENT OPTIONS IN ALLERGY 2022. [DOI: 10.1007/s40521-022-00319-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
19
|
Zhang K, Tan J, Hao X, Tang H, Abbas MN, Su J, Su Y, Cui H. Bombyx mori U-shaped regulates the melanization cascade and immune response via binding with the Lozenge protein. INSECT SCIENCE 2022; 29:704-716. [PMID: 34331739 DOI: 10.1111/1744-7917.12959] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Revised: 07/01/2021] [Accepted: 07/27/2021] [Indexed: 06/13/2023]
Abstract
Zinc finger protein, an important transcription factor, regulates gene expression associated with various physiological and pathological processes. U-shaped, belong to the Friend of GATA (FOG) transcription factor, plays a crucial role in hematopoiesis by interacting with the GATA transcription factor as a co-factor. However, little is known about its functions in insects. In the present study, a U-shaped cDNA was identified and characterized from the silkworm Bombyx mori and its potential roles in innate immunity investigated. The predicted silkworm U-shaped amino acid sequence contained a classical nuclear localization signal (NLS) motif "GESSPKRRRR" at position 450-459, and arginine residues at position 456 and 478 are the critical sites of the NLS. U-shaped mRNA was detected in all tested tissues of the B. mori; however, the highest levels were found in the hemocytes. U-shaped mRNA expression levels were upregulated in the hemocyte after the Escherichia coli and Staphylococcus aureus challenge. Furthermore, U-shaped knockdown significantly reduced the melanization process and suppressed the expression of melanization-associated genes, including PPO1, PPO2, PPAE and BAEE. In addition, U-shaped interacts with Lozenge protein to regulate the innate immune response of the insect. Our results revealed that U-shaped binds directly to Lozenge protein to modulate the melanization process and innate immune responses in silkworm.
Collapse
Affiliation(s)
- Kui Zhang
- State Key Laboratory of Silkworm Genome Biology, College of Sericulture, Textile and Biomass Sciences, Southwest University, Chongqing, 400716, China
| | - Juan Tan
- State Key Laboratory of Silkworm Genome Biology, College of Sericulture, Textile and Biomass Sciences, Southwest University, Chongqing, 400716, China
| | - Xiangwei Hao
- Chongqing Reproductive and Genetics Institute, Chongqing Obstetrics and Gynecology Hospital, Chongqing, 400013, China
| | - Houyi Tang
- State Key Laboratory of Silkworm Genome Biology, College of Sericulture, Textile and Biomass Sciences, Southwest University, Chongqing, 400716, China
| | - Muhammad Nadeem Abbas
- State Key Laboratory of Silkworm Genome Biology, College of Sericulture, Textile and Biomass Sciences, Southwest University, Chongqing, 400716, China
| | - Jingjing Su
- State Key Laboratory of Silkworm Genome Biology, College of Sericulture, Textile and Biomass Sciences, Southwest University, Chongqing, 400716, China
| | - Yongyue Su
- Department of Orthopaedic, 920th Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Kunming, 650032, China
| | - Hongjuan Cui
- State Key Laboratory of Silkworm Genome Biology, College of Sericulture, Textile and Biomass Sciences, Southwest University, Chongqing, 400716, China
- Cancer Center, Reproductive Medicine Center, Medical Research Institute, Southwest University, Chongqing, 400716, China
| |
Collapse
|
20
|
Jin X, Shao X, Pang W, Wang Z, Huang J. Sex-determining Region Y-box transcription factor 13 promotes breast cancer cell proliferation and glycolysis by activating the tripartite motif containing 11-mediated Wnt/β-catenin signaling pathway. Bioengineered 2022; 13:13033-13044. [PMID: 35611828 PMCID: PMC9276007 DOI: 10.1080/21655979.2022.2073127] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Breast cancer is the most frequent cancer among women and the second highest mortality in female across the world. Recent studies have illustrated that sex-determining region Y (SRY)-box protein (SOX) family plays essential roles in regulating various cancers. Nevertheless, the detailed effects of SOX13 on breast cancer are still uncovered. In our present study, SOX13 protein level was measured by using western blot assay in tissues and cells, and the results showed that SOX13 was upregulated in breast cancer tissues and cells compared with normal samples. Moreover, silencing SOX13 inhibited breast cancer cell viability, arrested cell cycle at G1/S phase and suppressed glycolysis, while overexpression of SOX13 reversed these events. Additionally, SOX13 knockdown reduced the level of proteins related to Wnt/β-catenin signaling pathway, whereas overexpression of tripartite motif containing 11 (TRM11) efficiently attenuated the effects, indicating that SOX13 controlled Wnt/β-catenin pathway depending on TRIM11. Furthermore, the data gained from xenograft tumor model illustrated that silencing SOX13 suppressed the tumor growth in nude mice and the glycolysis of tissues. In conclusion, our investigation illustrated that SOX13 facilitated breast cancer cell proliferation and glycolysis by modulating Wnt/β-catenin signaling pathway affected via TRIM11.
Collapse
Affiliation(s)
- Xiaoyan Jin
- Department of Breast Surgery, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China.,Department of Breast Surgery, Taizhou Municipal Hospital, Taizhou, Zhejiang Province, China
| | - Xuan Shao
- Department of Breast Surgery, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Wenyang Pang
- Department of Breast Surgery, Taizhou Municipal Hospital, Taizhou, Zhejiang Province, China
| | - Zhengyi Wang
- Department of Breast Surgery, Taizhou Municipal Hospital, Taizhou, Zhejiang Province, China
| | - Jian Huang
- Department of Breast Surgery, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| |
Collapse
|
21
|
Zhou Q, Yang HJ, Zuo MZ, Tao YL. Distinct expression and prognostic values of GATA transcription factor family in human ovarian cancer. J Ovarian Res 2022; 15:49. [PMID: 35488350 PMCID: PMC9052646 DOI: 10.1186/s13048-022-00974-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2020] [Accepted: 03/28/2022] [Indexed: 11/23/2022] Open
Abstract
Accumulated studies have provided controversial evidences of expression patterns and prognostic value of the GATA family in human ovarian cancer. In the present study, we accessed the distinct expression and prognostic roles of 7 individual members of GATA family in ovarian cancer (OC) patients through Oncomine analysis, CCLE analysis, Human Protein Atlas (HPA), Kaplan–Meier plotter (KM plotter) database, cBioPortal and Metascape. Our results indicated that GATA1, GATA3, GATA4 and TRPS1 mRNA and protein expression was significantly higher in OC than normal samples. High expression of GATA1, GATA2, and GATA4 were significantly correlated with better overall survival (OS), while increased GATA3 and GATA6 expression were associated with worse prognosis in OC patients. GATA1, GATA2, GATA3 and GATA6 were closely related to the different pathological histology, pathological grade, clinical stage and TP53 mutation status of OC. The genetic variation and interaction of the GATA family may be closely related to the pathogenesis and prognosis of OC, and the regulatory network composed of GATA family genes and their neighboring genes are mainly involved in Notch signaling pathway, Th1 and Th2 cell differentiation and Hippo signaling pathway. Transcriptional GATA1/2/3/4/6 could be prognostic markers and potential therapeutic target for OC patients.
Collapse
Affiliation(s)
- Quan Zhou
- Department of Gynecology and Obstetrics, the People's Hospital of China Three Gorges University/the First People's Hospital of Yichang, 2, Jie-fang Road, Yi chang, Yichang, 443000, Hubei, China.
| | - Huai-Jie Yang
- Department of Gynecology and Obstetrics, the People's Hospital of China Three Gorges University/the First People's Hospital of Yichang, 2, Jie-fang Road, Yi chang, Yichang, 443000, Hubei, China
| | - Man-Zhen Zuo
- Department of Gynecology and Obstetrics, the People's Hospital of China Three Gorges University/the First People's Hospital of Yichang, 2, Jie-fang Road, Yi chang, Yichang, 443000, Hubei, China
| | - Ya-Ling Tao
- Department of Gynecology and Obstetrics, the People's Hospital of China Three Gorges University/the First People's Hospital of Yichang, 2, Jie-fang Road, Yi chang, Yichang, 443000, Hubei, China
| |
Collapse
|
22
|
Liu J, Lin F, Wang X, Li C, Qi Q. GATA binding protein 5-mediated transcriptional activation of transmembrane protein 100 suppresses cell proliferation, migration and epithelial-to-mesenchymal transition in prostate cancer DU145 cells. Bioengineered 2022; 13:7972-7983. [PMID: 35358005 PMCID: PMC9162018 DOI: 10.1080/21655979.2021.2018979] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
It has been reported that transmembrane protein 100 (TMEM100) acts as a tumor regulator in several types of cancers. However, whether the expression of TMEM100 is associated with the development and prognosis of prostate cancer (PCa) remains elusive. Therefore, the present study aimed to uncover the role of GATA binding protein 5 (GATA5)-mediated activation of TMEM100 in the proliferation, migration and epithelial-to-mesenchymal transition (EMT) of PCa cells. The expressions of TMEM100 and GATA5 in PCa patients were analyzed by the GEPIA database. The binding site of GATA5 and TMEM100 promoter was predicted by the JASPAR database. Expressions of TMEM100 and GATA5 in PCa cells were detected by qRT-PCR and Western blot analysis. Cell Counting Kit 8 and colony formation assays were performed to measure cell proliferation. In addition, cell migration, invasion and the expression of EMT-associated proteins were evaluated using wound healing, transwell assay and Western blotting assays, respectively. The bioinformatics analysis revealed that TMEM100 was downregulated in PCa and was associated with overall survival of PCa. In addition, TMEM10 overexpression attenuated cell proliferation, migration, invasion and EMT in PCa cells. The interaction between TMEM100 and GATA5 was verified using dual luciferase reporter and chromatin immunoprecipitation assays. Furthermore, the results showed that GATA5 was downregulated and GATA5 silencing reversed the inhibitory effects of TMEM10 on PCa cells. Overall, the current study suggested that the GATA5-mediated transcriptional activation of TMEM100 could affect the behavior of PCa cells and was associated with poor prognosis in PCa.
Collapse
Affiliation(s)
- Jiaolin Liu
- Department of Urology, The Central Hospital of Linyi, Linyi, Shandong, China
| | - Fanlu Lin
- Department of Urology, The Central Hospital of Linyi, Linyi, Shandong, China
| | - Xin Wang
- Department of Urology, Linyi People's Hospital, Linyi, Shandong, China
| | - Chaopeng Li
- Department of Urology, The Central Hospital of Linyi, Linyi, Shandong, China
| | - Qiangyuan Qi
- Department of Urology, The Central Hospital of Linyi, Linyi, Shandong, China
| |
Collapse
|
23
|
Ding Q, Huo L, Peng Y, Yoon EC, Li Z, Sahin AA. Immunohistochemical Markers for Distinguishing Metastatic Breast Carcinoma from Other Common Malignancies: Update and Revisit. Semin Diagn Pathol 2022; 39:313-321. [DOI: 10.1053/j.semdp.2022.04.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 04/02/2022] [Accepted: 04/11/2022] [Indexed: 11/11/2022]
|
24
|
Zhang C, Zhao H, Song X, Wang J, Zhao S, Deng H, He L, Zhou X, Yin X, Zhang K, Zhang Y, Wu Z, Chen Q, Du J, Yu D, Zhang S, Deng W. Transcription factor GATA4 drives RNA polymerase III-directed transcription and transformed cell proliferation through a filamin A/GATA4/SP1 pathway. J Biol Chem 2022; 298:101581. [PMID: 35038452 PMCID: PMC8857480 DOI: 10.1016/j.jbc.2022.101581] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 12/31/2021] [Accepted: 01/03/2022] [Indexed: 11/27/2022] Open
Abstract
RNA polymerase III (pol III) products play fundamental roles in a variety of cellular processes, including protein synthesis and cancer cell proliferation. In addition, dysregulation of pol III-directed transcription closely correlates with tumorigenesis. It is therefore of interest to identify novel pathways or factors governing pol III-directed transcription. Here, we show that transcription factor (TF) GATA binding protein 4 (GATA4) expression in SaOS2 cells was stimulated by the silencing of filamin A (FLNA), a repressor of pol III-directed transcription, suggesting that GATA4 is potentially associated with the regulation of pol III-directed transcription. Indeed, we show that GATA4 expression positively correlates with pol III-mediated transcription and tumor cell proliferation. Mechanistically, we found that GATA4 depletion inhibits the occupancies of the pol III transcription machinery factors at the loci of pol III target genes by reducing expression of both TFIIIB subunit TFIIB-related factor 1 and TFIIIC subunit general transcription factor 3C subunit 2 (GTF3C2). GATA4 has been shown to activate specificity factor 1 (Sp1) gene transcription by binding to the Sp1 gene promoter, and Sp1 has been confirmed to activate pol III gene transcription by directly binding to both Brf1 and Gtf3c2 gene promoters. Thus, the findings from this study suggest that GATA4 links FLNA and Sp1 signaling to form an FLNA/GATA4/Sp1 axis to modulate pol III-directed transcription and transformed cell proliferation. Taken together, these results provide novel insights into the regulatory mechanism of pol III-directed transcription.
Collapse
Affiliation(s)
- Cheng Zhang
- School of Life Science and Health, Wuhan University of Science and Technology, Wuhan, China
| | - Houliang Zhao
- School of Life Science and Health, Wuhan University of Science and Technology, Wuhan, China
| | - Xiaoye Song
- School of Life Science and Health, Wuhan University of Science and Technology, Wuhan, China
| | - Juan Wang
- School of Materials and Metallurgy, Wuhan University of Science and Technology, Wuhan, China
| | - Shasha Zhao
- School of Life Science and Health, Wuhan University of Science and Technology, Wuhan, China
| | - Huan Deng
- School of Life Science and Health, Wuhan University of Science and Technology, Wuhan, China
| | - Liu He
- School of Life Science and Health, Wuhan University of Science and Technology, Wuhan, China
| | - Xiangyu Zhou
- School of Life Science and Health, Wuhan University of Science and Technology, Wuhan, China
| | - Xiaomei Yin
- School of Life Science and Health, Wuhan University of Science and Technology, Wuhan, China
| | - Kewei Zhang
- School of Life Science and Health, Wuhan University of Science and Technology, Wuhan, China
| | - Yue Zhang
- School of Life Science and Health, Wuhan University of Science and Technology, Wuhan, China
| | - Zhongyu Wu
- School of Life Science and Health, Wuhan University of Science and Technology, Wuhan, China
| | - Qiyue Chen
- School of Life Science and Health, Wuhan University of Science and Technology, Wuhan, China
| | - Jiannan Du
- School of Life Science and Health, Wuhan University of Science and Technology, Wuhan, China
| | - Deen Yu
- School of Life Science and Health, Wuhan University of Science and Technology, Wuhan, China
| | - Shihua Zhang
- School of Life Science and Health, Wuhan University of Science and Technology, Wuhan, China.
| | - Wensheng Deng
- School of Life Science and Health, Wuhan University of Science and Technology, Wuhan, China.
| |
Collapse
|
25
|
Lin CY, Kuo TT, Chang YC. Comparative Immunohistochemical Study of Hidroacanthoma Simplex and Clonal Seborrheic Keratosis With GATA3 and p63. Am J Dermatopathol 2022; 44:17-20. [PMID: 34132661 DOI: 10.1097/dad.0000000000001998] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
ABSTRACT Histopathologically both hidroacanthoma simplex (HS) and clonal seborrheic keratosis (CSK) are characterized by intraepidermal nests of tumor cells. Although they show subtle microscopic differences, they can be difficult to accurately differentiate. Previous immunohistochemical studies have been inconclusive. We conducted an immunohistochemical study with GATA3 and p63 on cases of HS and CSK tentatively identified by their microscopic appearances and cases of eccrine poroma and seborrheic keratosis as their respective controls. The clinical, histopathological, and dermoscopic findings of these cases were also reviewed. All cases of HS and poroma were negative for GATA3, whereas all cases of CSK and seborrheic keratosis were positive for GATA3. HS, CSK, and their controls were all positive for p63. Microscopic, clinical, and dermoscopic differences were also found between HS and CSK. Our study demonstrated that GATA3 is useful for differentiating HS from CSK. Our initial microscopic observations also proved to be reliable, but immunostaining with GATA3 is helpful for confirming the diagnosis or establishing the diagnosis of uncertain cases. Awareness of the clinical and dermoscopic features of these 2 entities could also avoid misdiagnosis based solely on pathological observation.
Collapse
Affiliation(s)
- Chien-Yio Lin
- Department of Dermatology, Chang Gung Memorial Hospital, Linkou and Taipei, Taiwan
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan; and
| | - Tseng-Tong Kuo
- Department of Dermatology, Chang Gung Memorial Hospital, Linkou and Taipei, Taiwan
| | - Ya-Ching Chang
- Department of Dermatology, Chang Gung Memorial Hospital, Linkou and Taipei, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
| |
Collapse
|
26
|
Leach DA, Fernandes RC, Bevan CL. Cellular specificity of androgen receptor, coregulators, and pioneer factors in prostate cancer. ENDOCRINE ONCOLOGY (BRISTOL, ENGLAND) 2022; 2:R112-R131. [PMID: 37435460 PMCID: PMC10259329 DOI: 10.1530/eo-22-0065] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 09/08/2022] [Indexed: 07/13/2023]
Abstract
Androgen signalling, through the transcription factor androgen receptor (AR), is vital to all stages of prostate development and most prostate cancer progression. AR signalling controls differentiation, morphogenesis, and function of the prostate. It also drives proliferation and survival in prostate cancer cells as the tumour progresses; given this importance, it is the main therapeutic target for disseminated disease. AR is also essential in the surrounding stroma, for the embryonic development of the prostate and controlling epithelial glandular development. Stromal AR is also important in cancer initiation, regulating paracrine factors that excite cancer cell proliferation, but lower stromal AR expression correlates with shorter time to progression/worse outcomes. The profile of AR target genes is different between benign and cancerous epithelial cells, between castrate-resistant prostate cancer cells and treatment-naïve cancer cells, between metastatic and primary cancer cells, and between epithelial cells and fibroblasts. This is also true of AR DNA-binding profiles. Potentially regulating the cellular specificity of AR binding and action are pioneer factors and coregulators, which control and influence the ability of AR to bind to chromatin and regulate gene expression. The expression of these factors differs between benign and cancerous cells, as well as throughout disease progression. The expression profile is also different between fibroblast and mesenchymal cell types. The functional importance of coregulators and pioneer factors in androgen signalling makes them attractive therapeutic targets, but given the contextual expression of these factors, it is essential to understand their roles in different cancerous and cell-lineage states.
Collapse
Affiliation(s)
- Damien A Leach
- Division of Cancer, Imperial Centre for Translational & Experimental Medicine, Imperial College London, Hammersmith Hospital Campus, London, UK
| | - Rayzel C Fernandes
- Division of Cancer, Imperial Centre for Translational & Experimental Medicine, Imperial College London, Hammersmith Hospital Campus, London, UK
| | - Charlotte L Bevan
- Division of Cancer, Imperial Centre for Translational & Experimental Medicine, Imperial College London, Hammersmith Hospital Campus, London, UK
| |
Collapse
|
27
|
Repurposing of the ALK Inhibitor Crizotinib for Acute Leukemia and Multiple Myeloma Cells. Pharmaceuticals (Basel) 2021; 14:ph14111126. [PMID: 34832908 PMCID: PMC8617756 DOI: 10.3390/ph14111126] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 10/28/2021] [Accepted: 11/02/2021] [Indexed: 12/18/2022] Open
Abstract
Crizotinib was a first generation of ALK tyrosine kinase inhibitor approved for the treatment of ALK-positive non-small-cell lung carcinoma (NSCLC) patients. COMPARE and cluster analyses of transcriptomic data of the NCI cell line panel indicated that genes with different cellular functions regulated the sensitivity or resistance of cancer cells to crizotinib. Transcription factor binding motif analyses in gene promoters divulged two transcription factors possibly regulating the expression of these genes, i.e., RXRA and GATA1, which are important for leukemia and erythroid development, respectively. COMPARE analyses also implied that cell lines of various cancer types displayed varying degrees of sensitivity to crizotinib. Unexpectedly, leukemia but not lung cancer cells were the most sensitive cells among the different types of NCI cancer cell lines. Re-examining this result in another panel of cell lines indeed revealed that crizotinib exhibited potent cytotoxicity towards acute myeloid leukemia and multiple myeloma cells. P-glycoprotein-overexpressing CEM/ADR5000 leukemia cells were cross-resistant to crizotinib. NCI-H929 multiple myeloma cells were the most sensitive cells. Hence, we evaluated the mode of action of crizotinib on these cells. Although crizotinib is a TKI, it showed highest correlation rates with DNA topoisomerase II inhibitors and tubulin inhibitors. The altered gene expression profiles after crizotinib treatment predicted several networks, where TOP2A and genes related to cell cycle were downregulated. Cell cycle analyses showed that cells incubated with crizotinib for 24 h accumulated in the G2M phase. Crizotinib also increased the number of p-H3(Ser10)-positive NCI-H929 cells illustrating crizotinib's ability to prevent mitotic exit. However, cells accumulated in the sub-G0G1 fraction with longer incubation periods, indicating apoptosis induction. Additionally, crizotinib disassembled the tubulin network of U2OS cells expressing an α-tubulin-GFP fusion protein, preventing migration of cancer cells. This result was verified by in vitro tubulin polymerization assays. In silico molecular docking also revealed a strong binding affinity of crizotinib to the colchicine and Vinca alkaloid binding sites. Taken together, these results demonstrate that crizotinib destabilized microtubules. Additionally, the decatenation assay showed that crizotinib partwise inhibited the catalytic activity of DNA topoisomerase II. In conclusion, crizotinib exerted kinase-independent cytotoxic effects through the dual inhibition of tubulin polymerization and topoisomerase II and might be used to treat not only NSCLC but also multiple myeloma.
Collapse
|
28
|
Exploring the Inflammatory Pathogenesis of Colorectal Cancer. Diseases 2021; 9:diseases9040079. [PMID: 34842660 PMCID: PMC8628792 DOI: 10.3390/diseases9040079] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 10/21/2021] [Accepted: 10/23/2021] [Indexed: 12/12/2022] Open
Abstract
Colorectal cancer is one of the most commonly diagnosed cancers worldwide. Traditionally, mechanisms of colorectal cancer formation have focused on genetic alterations including chromosomal damage and microsatellite instability. In recent years, there has been a growing body of evidence supporting the role of inflammation in colorectal cancer formation. Multiple cytokines, immune cells such T cells and macrophages, and other immune mediators have been identified in pathways leading to the initiation, growth, and metastasis of colorectal cancer. Outside the previously explored mechanisms and pathways leading to colorectal cancer, initiatives have been shifted to further study the role of inflammation in pathogenesis. Inflammatory pathways have also been linked to some traditional risk factors of colorectal cancer such as obesity, smoking and diabetes, as well as more novel associations such as the gut microbiome, the gut mycobiome and exosomes. In this review, we will explore the roles of obesity and diet, smoking, diabetes, the microbiome, the mycobiome and exosomes in colorectal cancer, with a specific focus on the underlying inflammatory and metabolic pathways involved. We will also investigate how the study of colon cancer from an inflammatory background not only creates a more holistic and inclusive understanding of this disease, but also creates unique opportunities for prevention, early diagnosis and therapy.
Collapse
|
29
|
Role of GATA3 in tumor diagnosis: A review. Pathol Res Pract 2021; 226:153611. [PMID: 34547599 DOI: 10.1016/j.prp.2021.153611] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 09/05/2021] [Accepted: 09/08/2021] [Indexed: 11/21/2022]
Abstract
GATA binding protein 3 (GATA3) belongs to a family of transcription factors comprising six members. These proteins identify G-A-T-A containing sequences in the target gene and bind to DNA target via two zinc-finger domains. The aim of this study was to evaluate the role of GATA3 in the diagnosis of tumors and its value as a prognostic marker. To perform this review, a comprehensive search was conducted through PubMed, Embase, Scopus, Cochrane and Google Scholar databases from 1985 to 2020. Articles were considered thoroughly by independent reviewers and data were extracted in predefined forms. Final synthesis was conducted by using appropriate data from included articles in each topic. Studies have shown that GATA3 has a critical role in the development of epithelial structures in both embryonic and adult tissues. The majority of studies regarding GATA3 expression in tumor evaluation focused on breast and urothelial neoplasms, whether primary or metastatic. Its sensitivity in these neoplasms has been reported to be high and made this marker more valuable than other available immunohistochemistry markers. However, GATA3 expression was not restricted to these tumors. Studies have shown that GATA3 immunostaining could be a useful tool in various tumors in kidney, salivary gland, endocrine system, hematopoietic system, and skin. GATA3 can also be used as a useful prognostic tool. Although GATA3 is a multi-specific immunohistochemical stain, it is a valuable marker in the panel for confirming many epithelial or mesenchymal neoplasms as both a diagnostic and prognostic tool.
Collapse
|
30
|
Ong SLM, Szuhai K, Bovée JVMG. Gene fusions in vascular tumors and their underlying molecular mechanisms. Expert Rev Mol Diagn 2021; 21:897-909. [PMID: 34225547 DOI: 10.1080/14737159.2021.1950533] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
INTRODUCTION The group of vascular tumors contains many different entities, and is considered difficult by pathologists, as they often have overlapping histological characteristics. Chromosomal translocations have been identified in ~20% of mesenchymal tumors and are considered the drivers of tumor formation. Many translocations have been discovered over the past decade through next-generation sequencing. This technological advancement has also revealed several recurrent gene fusions in vascular tumors. AREAS COVERED This review will discuss the various vascular tumors for which recurrent gene fusions have been identified. The gene fusions and the presumed molecular mechanisms underlying tumorigenesis are shown, and potential implications for targeted therapies discussed. The identification of these gene fusions in vascular tumors has improved diagnostic accuracy, especially since several of these fusions can be easily detected using surrogate immunohistochemical markers. EXPERT OPINION The identification of gene fusions in a subset of vascular tumors over the past decade has improved diagnostic accuracy, and has provided the pathologists with novel diagnostic tools to accurately diagnose these often difficult tumors. Moreover, the increased understanding of the underlying molecular mechanisms can guide the development of targeted therapeutic strategies.
Collapse
Affiliation(s)
- Sheena L M Ong
- Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands
| | - Karoly Szuhai
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, The Netherlands
| | - Judith V M G Bovée
- Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
31
|
Breton JM, Arkun K, Tischler AS, Qamar AS, Sillman JS, Heilman CB. Clinical and histopathological principles for the diagnosis of a recurrent paraganglioma of the jugular foramen initially diagnosed as a middle ear adenoma: illustrative case. JOURNAL OF NEUROSURGERY: CASE LESSONS 2021; 2:CASE21307. [PMID: 36131578 PMCID: PMC9563646 DOI: 10.3171/case21307] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 05/20/2021] [Indexed: 01/16/2023]
Abstract
BACKGROUND Paragangliomas (PGLs) are rare neoplasms that may be associated with hereditary PGL syndromes and variable risk of metastasis. Middle ear adenomas are extremely rare tumors with no known hereditary predisposition and extremely low risk of metastasis. Although often easily differentiated, they may share clinical and pathological features that misdirect and confuse the diagnosis. OBSERVATIONS The authors discussed a 35-year-old woman with left-sided hearing loss and bleeding from the external ear canal who presented to an outside hospital. She underwent resection of a middle ear and mastoid mass, initially diagnosed as a middle ear adenoma with neuroendocrine features, with later mastoidectomy and ligation of the sigmoid sinus with microsurgical excision of persistent tumor in the jugular foramen and temporal bone. Histopathologically, her tumor was vascular, composed of benign-appearing epithelioid cells with “salt and pepper” neuroendocrine chromatin arranged in vague nests. Lesional cells were GATA3-immunopositive, glucagon-negative, and succinate dehydrogenase-immunonegative, consistent with PGL rather than middle ear adenoma, and required further workup for hereditary PGL syndromes. LESSONS This case demonstrates potential challenges in differentiating a PGL from a middle ear adenoma. The authors offer clinical, histopathological, and imaging principles to aid in diagnosis and workup.
Collapse
Affiliation(s)
- Jeffrey M. Breton
- Department of Neurosurgery, Tufts Medical Center and Tufts University School of Medicine, Boston, Massachusetts; and
| | - Knarik Arkun
- Department of Neurosurgery, Tufts Medical Center and Tufts University School of Medicine, Boston, Massachusetts; and
- Departments of Pathology and Laboratory Medicine and
| | | | | | | | - Carl B. Heilman
- Department of Neurosurgery, Tufts Medical Center and Tufts University School of Medicine, Boston, Massachusetts; and
| |
Collapse
|
32
|
Network pharmacology of triptolide in cancer cells: implications for transcription factor binding. Invest New Drugs 2021; 39:1523-1537. [PMID: 34213719 PMCID: PMC8541937 DOI: 10.1007/s10637-021-01137-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Accepted: 06/10/2021] [Indexed: 01/29/2023]
Abstract
Background Triptolide is an active natural product, which inhibits cell proliferation, induces cell apoptosis, suppresses tumor metastasis and improves the effect of other therapeutic treatments in several cancer cell lines by affecting multiple molecules and signaling pathways, such as caspases, heat-shock proteins, DNA damage and NF-ĸB. Purpose We investigated the effect of triptolide towards NF-ĸB and GATA1. Methods We used cell viability assay, compare and cluster analyses of microarray-based mRNA transcriptome-wide expression data, gene promoter binding motif analysis, molecular docking, Ingenuity pathway analysis, NF-ĸB reporter cell assay, and electrophoretic mobility shift assay (EMSA) of GATA1. Results Triptolide inhibited the growth of drug-sensitive (CCRF-CEM, U87.MG) and drug-resistant cell lines (CEM/ADR5000, U87.MGΔEGFR). Hierarchical cluster analysis showed six major clusters in dendrogram. The sensitive and resistant cell lines were statistically significant (p = 0.65 × 10-2) distributed. The binding motifs of NF-κB (Rel) and of GATA1 proteins were significantly enriched in regions of 25 kb upstream promoter of all genes. IPA showed the networks, biological functions, and canonical pathways influencing the activity of triptolide towards tumor cells. Interestingly, upstream analysis for the 40 genes identified by compare analysis revealed ZFPM1 (friend of GATA protein 1) as top transcription regulator. However, we did not observe any effect of triptolide to the binding of GATA1 in vitro. We confirmed that triptolide inhibited NF-κB activity, and it strongly bound to the pharmacophores of IκB kinase β and NF-κB in silico. Conclusion Triptolide showed promising inhibitory effect toward NF-κB, making it a potential candidate for targeting NF-κB.
Collapse
|
33
|
Islam Z, Ali AM, Naik A, Eldaw M, Decock J, Kolatkar PR. Transcription Factors: The Fulcrum Between Cell Development and Carcinogenesis. Front Oncol 2021; 11:681377. [PMID: 34195082 PMCID: PMC8236851 DOI: 10.3389/fonc.2021.681377] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 05/26/2021] [Indexed: 12/15/2022] Open
Abstract
Higher eukaryotic development is a complex and tightly regulated process, whereby transcription factors (TFs) play a key role in controlling the gene regulatory networks. Dysregulation of these regulatory networks has also been associated with carcinogenesis. Transcription factors are key enablers of cancer stemness, which support the maintenance and function of cancer stem cells that are believed to act as seeds for cancer initiation, progression and metastasis, and treatment resistance. One key area of research is to understand how these factors interact and collaborate to define cellular fate during embryogenesis as well as during tumor development. This review focuses on understanding the role of TFs in cell development and cancer. The molecular mechanisms of cell fate decision are of key importance in efforts towards developing better protocols for directed differentiation of cells in research and medicine. We also discuss the dysregulation of TFs and their role in cancer progression and metastasis, exploring TF networks as direct or indirect targets for therapeutic intervention, as well as specific TFs' potential as biomarkers for predicting and monitoring treatment responses.
Collapse
Affiliation(s)
- Zeyaul Islam
- Diabetes Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation, Doha, Qatar
| | - Ameena Mohamed Ali
- Diabetes Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation, Doha, Qatar
| | - Adviti Naik
- Translational Cancer and Immunity Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation, Doha, Qatar
| | - Mohamed Eldaw
- Diabetes Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation, Doha, Qatar
| | - Julie Decock
- Translational Cancer and Immunity Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation, Doha, Qatar
| | - Prasanna R. Kolatkar
- Diabetes Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation, Doha, Qatar
| |
Collapse
|
34
|
Serag Eldien MM, Abdou AG, Elghrabawy GRA, Alhanafy AM, Mahmoud SF. Stratification of urothelial bladder carcinoma depending on immunohistochemical expression of GATA3 and CK5/6. J Immunoassay Immunochem 2021; 42:662-678. [PMID: 34106817 DOI: 10.1080/15321819.2021.1937212] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Bladder urothelial carcinoma (BUC) has two pathways with distinct molecular features and prognosis, non-muscle invasive (NMI) and muscle invasive (MI) tumors. The aim is to investigate the expression of GATA3 and CK5/6 in BUC with correlation to clinicopathologic parameters, including their impact on survival beside their potential use to stratify cases into prognostic subgroups. This study included 80 cases of BUC stained immunohistochemically by GATA3 and CK5/6. The cases were divided into four groups regarding expression status of both markers (luminal, basal, mixed, and null). GATA3 percentage of expression decreased in urothelial carcinoma with squamous differentiation, MI tumors, high-grade tumors, tumors with involved lymph nodes, presence of perineural invasion, presence of bilharziasis, presence of lympho-vascular invasion, and high mitotic count. CK5/6 positivity was higher in urothelial carcinoma cases with squamous differentiation, MI tumors, and presence of perineural invasion. Pure urothelial carcinoma and NMI were in favor of luminal group (GATA3 +ve/CK5/6 -ve). Univariate analysis showed that the presence of bilharziasis was associated with shorter PFS (p = .04). GATA3 and CK5/6 could be used for the stratification of urothelial bladder carcinoma into subtypes with different characteristics. Luminal bladder cancer represents the most common type (60%) that carries favorable features. Bilharziasis-associated urothelial carcinoma carries poor outcome manifested by short PFS.
Collapse
Affiliation(s)
| | - Asmaa Gaber Abdou
- Pathology Department, Faculty of Medicine, Menoufia University, Shebein Elkom, Egypt
| | | | - Alshimaa Mahmoud Alhanafy
- Clinical Oncology and Nuclear Medicine Department, Faculty of Medicine, Menoufia University, Shebein Elkom, Egypt
| | - Shereen Fathy Mahmoud
- Pathology Department, Faculty of Medicine, Menoufia University, Shebein Elkom, Egypt
| |
Collapse
|
35
|
DeLaForest A, Kohlnhofer BM, Franklin OD, Stavniichuk R, Thompson CA, Pulakanti K, Rao S, Battle MA. GATA4 Controls Epithelial Morphogenesis in the Developing Stomach to Promote Establishment of Glandular Columnar Epithelium. Cell Mol Gastroenterol Hepatol 2021; 12:1391-1413. [PMID: 34111600 PMCID: PMC8479485 DOI: 10.1016/j.jcmgh.2021.05.021] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2020] [Revised: 05/12/2021] [Accepted: 05/19/2021] [Indexed: 02/04/2023]
Abstract
BACKGROUND & AIMS The transcription factor GATA4 is broadly expressed in nascent foregut endoderm. As development progresses, GATA4 is lost in the domain giving rise to the stratified squamous epithelium of the esophagus and forestomach (FS), while it is maintained in the domain giving rise to the simple columnar epithelium of the hindstomach (HS). Differential GATA4 expression within these domains coincides with the onset of distinct tissue morphogenetic events, suggesting a role for GATA4 in diversifying foregut endoderm into discrete esophageal/FS and HS epithelial tissues. The goal of this study was to determine how GATA4 regulates differential morphogenesis of the mouse gastric epithelium. METHODS We used a Gata4 conditional knockout mouse line to eliminate GATA4 in the developing HS and a Gata4 conditional knock-in mouse line to express GATA4 in the developing FS. RESULTS We found that GATA4-deficient HS epithelium adopted a FS-like fate, and conversely, that GATA4-expressing FS epithelium adopted a HS-like fate. Underlying structural changes in these epithelia were broad changes in gene expression networks attributable to GATA4 directly activating or repressing expression of HS or FS defining transcripts. Our study implicates GATA4 as having a primary role in suppressing an esophageal/FS transcription factor network during HS development to promote columnar epithelium. Moreover, GATA4-dependent phenotypes in developmental mutants reflected changes in gene expression associated with Barrett's esophagus. CONCLUSIONS This study demonstrates that GATA4 is necessary and sufficient to activate the development of simple columnar epithelium, rather than stratified squamous epithelium, in the embryonic stomach. Moreover, similarities between mutants and Barrett's esophagus suggest that developmental biology can provide insight into human disease mechanisms.
Collapse
Affiliation(s)
- Ann DeLaForest
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Bridget M Kohlnhofer
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Olivia D Franklin
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Roman Stavniichuk
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Cayla A Thompson
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Kirthi Pulakanti
- Blood Research Institute, Versiti Wisconsin, Milwaukee, Wisconsin
| | - Sridhar Rao
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, Wisconsin; Blood Research Institute, Versiti Wisconsin, Milwaukee, Wisconsin; Division of Hematology/Oncology/Blood and Marrow Transplantation, Department of Pediatrics, Medical College of Wisconsin and Children's Wisconsin, Milwaukee, Wisconsin
| | - Michele A Battle
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, Wisconsin.
| |
Collapse
|
36
|
Shalmani A, Ullah U, Muhammad I, Zhang D, Sharif R, Jia P, Saleem N, Gul N, Rakhmanova A, Tahir MM, Chen KM, An N. The TAZ domain-containing proteins play important role in the heavy metals stress biology in plants. ENVIRONMENTAL RESEARCH 2021; 197:111030. [PMID: 33774015 DOI: 10.1016/j.envres.2021.111030] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 03/11/2021] [Accepted: 03/13/2021] [Indexed: 06/12/2023]
Abstract
TAZ (transcriptional coactivator with PDZ-binding) zinc finger domains, also known as transcription adaptor putative zinc finger domains, that control diverse function in plant growth and development. Here, in the present study, we evaluated the role of the TAZ domain-containing gene in response to various heavy metals. Initially, we found a total of 3, 7, 8, 9, 9, 9, 7, 14, 6, 10, and 6 proteins containing TAZ domain in stiff brome, millet, sorghum, potato, pepper, maize, rice, apple, peach, pear, and tomato genome that could trigger the plant resistance against various heavy metals, respectively. Various in-silico approaches were applied such as duplication, phylogenetic analysis, and gene structure, to understand the basic features of the TAZ domain-containing genes in plants. Gene expression analyses were also performed under heavy metals (Cr, Zn, Ni, Cd, Co, Fe, Mn, and Pb). The results of quantitative real-time PCR analysis indicated that the TAZ gene family members were differentially expressed under different heavy metals. We further characterized the functions of the TAZ domain-containing gene under the heavy metal stresses by overexpressing the OsTAZ4 gene in Arabidopsis. The TAZ genes could promote plant resistance against various heavy metals by interacting with OsMYB34 and OsFHA9 transcription factors. The results will contribute to elucidate the relationship of TAZ proteins with heavy metals stresses and also ascertain the biological function in plant growth and development.
Collapse
Affiliation(s)
- Abdullah Shalmani
- College of Horticulture, Northwest A and F University, Yangling, Shaanxi Province, 712100, China; State Key Laboratory of Crop Stress Biology in Arid Areas, College of Life Sciences, Northwest A&F University, Yangling, Shaanxi Province, 712100, China.
| | - Uzair Ullah
- Department of Genetics, Hazara University, Manshera, KPK, Pakistan.
| | - Izhar Muhammad
- State Key Laboratory of Crop Stress Biology in Arid Areas, College of Life Sciences, Northwest A&F University, Yangling, Shaanxi Province, 712100, China; College of Agronomy, Northwest A and F University, Yangling, Shaanxi Province, 712100, China.
| | - Dong Zhang
- College of Horticulture, Northwest A and F University, Yangling, Shaanxi Province, 712100, China.
| | - Rahat Sharif
- Department of Horticulture, School of Horticulture and Plant Protection, Yangzhou University, 48 Wenhui East Road, Yangzhou, Jiangsu, 225009, PR China.
| | - Peng Jia
- College of Horticulture, Northwest A and F University, Yangling, Shaanxi Province, 712100, China.
| | - Noor Saleem
- College of Agronomy, Northwest A and F University, Yangling, Shaanxi Province, 712100, China.
| | - Nazish Gul
- Department of Genetics, Hazara University, Manshera, KPK, Pakistan.
| | - Aizhan Rakhmanova
- College of Food Science and Engineering, Northwest A & F University, Yangling, Shaanxi Province, 712100, China.
| | - Muhammad Mobeen Tahir
- College of Horticulture, Northwest A and F University, Yangling, Shaanxi Province, 712100, China.
| | - Kun-Ming Chen
- State Key Laboratory of Crop Stress Biology in Arid Areas, College of Life Sciences, Northwest A&F University, Yangling, Shaanxi Province, 712100, China.
| | - Na An
- College of Horticulture, Northwest A and F University, Yangling, Shaanxi Province, 712100, China; College of Life Sciences, Northwest A&F University, Yangling, Shaanxi Province, 712100, China.
| |
Collapse
|
37
|
Heslop JA, Pournasr B, Liu JT, Duncan SA. GATA6 defines endoderm fate by controlling chromatin accessibility during differentiation of human-induced pluripotent stem cells. Cell Rep 2021; 35:109145. [PMID: 34010638 PMCID: PMC8202205 DOI: 10.1016/j.celrep.2021.109145] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 01/20/2021] [Accepted: 04/26/2021] [Indexed: 02/07/2023] Open
Abstract
In addition to driving specific gene expression profiles, transcriptional regulators are becoming increasingly recognized for their capacity to modulate chromatin structure. GATA6 is essential for the formation of definitive endoderm; however, the molecular basis defining the importance of GATA6 to endoderm commitment is poorly understood. The members of the GATA family of transcription factors have the capacity to bind and alter the accessibility of chromatin. Using pluripotent stem cells as a model of human development, we reveal that GATA6 is integral to the establishment of the endoderm enhancer network via the induction of chromatin accessibility and histone modifications. We additionally identify the chromatin-modifying complexes that interact with GATA6, defining the putative mechanisms by which GATA6 modulates chromatin architecture. The identified GATA6-dependent processes further our knowledge of the molecular mechanisms that underpin cell-fate decisions during formative development.
Collapse
Affiliation(s)
- James A Heslop
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, USA
| | - Behshad Pournasr
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, USA
| | - Jui-Tung Liu
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, USA
| | - Stephen A Duncan
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, USA.
| |
Collapse
|
38
|
Dölle A, Adhikari B, Krämer A, Weckesser J, Berner N, Berger LM, Diebold M, Szewczyk MM, Barsyte-Lovejoy D, Arrowsmith CH, Gebel J, Löhr F, Dötsch V, Eilers M, Heinzlmeir S, Kuster B, Sotriffer C, Wolf E, Knapp S. Design, Synthesis, and Evaluation of WD-Repeat-Containing Protein 5 (WDR5) Degraders. J Med Chem 2021; 64:10682-10710. [PMID: 33980013 DOI: 10.1021/acs.jmedchem.1c00146] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Histone H3K4 methylation serves as a post-translational hallmark of actively transcribed genes and is introduced by histone methyltransferase (HMT) and its regulatory scaffolding proteins. One of these is the WD-repeat-containing protein 5 (WDR5) that has also been associated with controlling long noncoding RNAs and transcription factors including MYC. The wide influence of dysfunctional HMT complexes and the typically upregulated MYC levels in diverse tumor types suggested WDR5 as an attractive drug target. Indeed, protein-protein interface inhibitors for two protein interaction interfaces on WDR5 have been developed. While such compounds only inhibit a subset of WDR5 interactions, chemically induced proteasomal degradation of WDR5 might represent an elegant way to target all oncogenic functions. This study presents the design, synthesis, and evaluation of two diverse WDR5 degrader series based on two WIN site binding scaffolds and shows that linker nature and length strongly influence degradation efficacy.
Collapse
Affiliation(s)
- Anja Dölle
- Structural Genomics Consortium (SGC), Buchmann Institute for Life Sciences (BMLS), Goethe University Frankfurt am Main, Max-von-Laue-Str. 15, 60438 Frankfurt am Main, Germany.,Institut für Pharmazeutische Chemie, Goethe University Frankfurt am Main, Max-von-Laue-Str. 9, 60438 Frankfurt am Main, Germany
| | - Bikash Adhikari
- Cancer Systems Biology Group, Theodor Boveri Institute, University of Würzburg, Am Hubland, 97074 Würzburg, Germany
| | - Andreas Krämer
- Structural Genomics Consortium (SGC), Buchmann Institute for Life Sciences (BMLS), Goethe University Frankfurt am Main, Max-von-Laue-Str. 15, 60438 Frankfurt am Main, Germany.,Institut für Pharmazeutische Chemie, Goethe University Frankfurt am Main, Max-von-Laue-Str. 9, 60438 Frankfurt am Main, Germany
| | - Janik Weckesser
- Structural Genomics Consortium (SGC), Buchmann Institute for Life Sciences (BMLS), Goethe University Frankfurt am Main, Max-von-Laue-Str. 15, 60438 Frankfurt am Main, Germany.,Institut für Pharmazeutische Chemie, Goethe University Frankfurt am Main, Max-von-Laue-Str. 9, 60438 Frankfurt am Main, Germany
| | - Nicola Berner
- Chair of Proteomics and Bioanalytics, Technical University of Munich, 85354 Freising, Germany.,German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Lena-Marie Berger
- Structural Genomics Consortium (SGC), Buchmann Institute for Life Sciences (BMLS), Goethe University Frankfurt am Main, Max-von-Laue-Str. 15, 60438 Frankfurt am Main, Germany.,Institut für Pharmazeutische Chemie, Goethe University Frankfurt am Main, Max-von-Laue-Str. 9, 60438 Frankfurt am Main, Germany
| | - Mathias Diebold
- Institut für Pharmazie und Lebensmittelchemie, University of Würzburg, Am Hubland, 97074 Würzburg, Germany
| | - Magdalena M Szewczyk
- Structural Genomics Consortium, University of Toronto, Toronto, ON M5G 1L7, Canada
| | - Dalia Barsyte-Lovejoy
- Structural Genomics Consortium, University of Toronto, Toronto, ON M5G 1L7, Canada.,Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON M5G 2M9, Canada
| | - Cheryl H Arrowsmith
- Structural Genomics Consortium, University of Toronto, Toronto, ON M5G 1L7, Canada.,Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2C1, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 2M9, Canada
| | - Jakob Gebel
- Structural Genomics Consortium (SGC), Buchmann Institute for Life Sciences (BMLS), Goethe University Frankfurt am Main, Max-von-Laue-Str. 15, 60438 Frankfurt am Main, Germany.,Institute of Biophysical Chemistry and Center for Biomolecular Magnetic Resonance and Cluster of Excellence Macromolecular Complexes (CEF), Goethe University Frankfurt am Main, 60438 Frankfurt am Main, Germany
| | - Frank Löhr
- Structural Genomics Consortium (SGC), Buchmann Institute for Life Sciences (BMLS), Goethe University Frankfurt am Main, Max-von-Laue-Str. 15, 60438 Frankfurt am Main, Germany.,Institute of Biophysical Chemistry and Center for Biomolecular Magnetic Resonance and Cluster of Excellence Macromolecular Complexes (CEF), Goethe University Frankfurt am Main, 60438 Frankfurt am Main, Germany
| | - Volker Dötsch
- Structural Genomics Consortium (SGC), Buchmann Institute for Life Sciences (BMLS), Goethe University Frankfurt am Main, Max-von-Laue-Str. 15, 60438 Frankfurt am Main, Germany.,Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 2M9, Canada
| | - Martin Eilers
- Department of Biochemistry and Molecular Biology, Theodor Boveri Institute, University of Würzburg, Am Hubland, 97074 Würzburg, Germany
| | - Stephanie Heinzlmeir
- Chair of Proteomics and Bioanalytics, Technical University of Munich, 85354 Freising, Germany
| | - Bernhard Kuster
- Chair of Proteomics and Bioanalytics, Technical University of Munich, 85354 Freising, Germany.,German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany.,Bavarian Biomolecular Mass Spectrometry Center (BayBioMS), Technical University of Munich, 85354 Freising, Germany
| | - Christoph Sotriffer
- Institut für Pharmazie und Lebensmittelchemie, University of Würzburg, Am Hubland, 97074 Würzburg, Germany
| | - Elmar Wolf
- Cancer Systems Biology Group, Theodor Boveri Institute, University of Würzburg, Am Hubland, 97074 Würzburg, Germany
| | - Stefan Knapp
- Structural Genomics Consortium (SGC), Buchmann Institute for Life Sciences (BMLS), Goethe University Frankfurt am Main, Max-von-Laue-Str. 15, 60438 Frankfurt am Main, Germany.,Institut für Pharmazeutische Chemie, Goethe University Frankfurt am Main, Max-von-Laue-Str. 9, 60438 Frankfurt am Main, Germany.,German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| |
Collapse
|
39
|
Davenport CF, Scheithauer T, Dunst A, Bahr FS, Dorda M, Wiehlmann L, Tran DDH. Genome-Wide Methylation Mapping Using Nanopore Sequencing Technology Identifies Novel Tumor Suppressor Genes in Hepatocellular Carcinoma. Int J Mol Sci 2021; 22:ijms22083937. [PMID: 33920410 PMCID: PMC8069345 DOI: 10.3390/ijms22083937] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 04/08/2021] [Accepted: 04/09/2021] [Indexed: 12/29/2022] Open
Abstract
Downregulation of multiple tumor suppressor genes (TSGs) plays an important role in cancer formation. Recent evidence has accumulated that cancer progression involves genome-wide alteration of epigenetic modifications, which may cause downregulation of the tumor suppressor gene. Using hepatocellular carcinoma (HCC) as a system, we mapped 5-methylcytosine signal at a genome-wide scale using nanopore sequencing technology to identify novel TSGs. Integration of methylation data with gene transcription profile of regenerated liver and primary HCCs allowed us to identify 10 potential tumor suppressor gene candidates. Subsequent validation led us to focus on functionally characterizing one candidate—glucokinase (GCK). We show here that overexpression of GCK inhibits the proliferation of HCC cells via induction of intracellular lactate accumulation and subsequently causes energy crisis due to NAD+ depletion. This suggests GCK functions as a tumor suppressor gene and may be involved in HCC development. In conclusion, these data provide valuable clues for further investigations of the process of tumorigenesis in human cancer.
Collapse
Affiliation(s)
- Colin F. Davenport
- Research Core Unit Genomics OE 9415, Medizinische Hochschule Hannover, Carl-Neuberg-Str. 1, D-30623 Hannover, Germany; (C.F.D.); (T.S.); (M.D.); (L.W.)
| | - Tobias Scheithauer
- Research Core Unit Genomics OE 9415, Medizinische Hochschule Hannover, Carl-Neuberg-Str. 1, D-30623 Hannover, Germany; (C.F.D.); (T.S.); (M.D.); (L.W.)
| | - Alessia Dunst
- Institut fuer Zellbiochemie, OE4310, Medizinische Hochschule Hannover, Carl-Neuberg-Str. 1, D-30623 Hannover, Germany; (A.D.); (F.S.B.)
| | - Frauke Sophie Bahr
- Institut fuer Zellbiochemie, OE4310, Medizinische Hochschule Hannover, Carl-Neuberg-Str. 1, D-30623 Hannover, Germany; (A.D.); (F.S.B.)
| | - Marie Dorda
- Research Core Unit Genomics OE 9415, Medizinische Hochschule Hannover, Carl-Neuberg-Str. 1, D-30623 Hannover, Germany; (C.F.D.); (T.S.); (M.D.); (L.W.)
| | - Lutz Wiehlmann
- Research Core Unit Genomics OE 9415, Medizinische Hochschule Hannover, Carl-Neuberg-Str. 1, D-30623 Hannover, Germany; (C.F.D.); (T.S.); (M.D.); (L.W.)
| | - Doan Duy Hai Tran
- Institut fuer Zellbiochemie, OE4310, Medizinische Hochschule Hannover, Carl-Neuberg-Str. 1, D-30623 Hannover, Germany; (A.D.); (F.S.B.)
- Correspondence: ; Tel.: +49-511-532-2857; Fax: +49-511-532-2847
| |
Collapse
|
40
|
Sivasudhan E, Blake N, Lu ZL, Meng J, Rong R. Dynamics of m6A RNA Methylome on the Hallmarks of Hepatocellular Carcinoma. Front Cell Dev Biol 2021; 9:642443. [PMID: 33869193 PMCID: PMC8047153 DOI: 10.3389/fcell.2021.642443] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 02/23/2021] [Indexed: 12/19/2022] Open
Abstract
Epidemiological data consistently rank hepatocellular carcinoma (HCC) as one of the leading causes of cancer-related deaths worldwide, often posing severe economic burden on health care. While the molecular etiopathogenesis associated with genetic and epigenetic modifications has been extensively explored, the biological influence of the emerging field of epitranscriptomics and its associated aberrant RNA modifications on tumorigenesis is a largely unexplored territory with immense potential for discovering new therapeutic approaches. In particular, the underlying cellular mechanisms of different hallmarks of hepatocarcinogenesis that are governed by the complex dynamics of m6A RNA methylation demand further investigation. In this review, we reveal the up-to-date knowledge on the mechanistic and functional link between m6A RNA methylation and pathogenesis of HCC.
Collapse
Affiliation(s)
- Enakshi Sivasudhan
- Department of Biological Sciences, Xi'an Jiaotong-Liverpool University, Suzhou, China.,Department of Clinical Infection, Microbiology and Immunology, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Neil Blake
- Department of Clinical Infection, Microbiology and Immunology, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Zhi-Liang Lu
- Department of Biological Sciences, Xi'an Jiaotong-Liverpool University, Suzhou, China.,Institute of Integrative Biology, University of Liverpool, Liverpool, United Kingdom
| | - Jia Meng
- Department of Biological Sciences, Xi'an Jiaotong-Liverpool University, Suzhou, China.,Institute of Integrative Biology, University of Liverpool, Liverpool, United Kingdom
| | - Rong Rong
- Department of Biological Sciences, Xi'an Jiaotong-Liverpool University, Suzhou, China.,Institute of Integrative Biology, University of Liverpool, Liverpool, United Kingdom
| |
Collapse
|
41
|
Jin C, Hacking S, Sajjan S, Kamanda S, Bhuiya T, Nasim M. GATA binding protein 3 (GATA3) as a marker for metaplastic spindle cell carcinoma of the breast. Pathol Res Pract 2021; 221:153413. [PMID: 33770555 DOI: 10.1016/j.prp.2021.153413] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 03/14/2021] [Accepted: 03/16/2021] [Indexed: 10/21/2022]
Abstract
Spindle cell lesions of the breast comprise a diverse set of tumors; harboring significant histological and immunohistochemical (IHC) overlap. Accurate diagnosis and classification of spindle cell lesions in the breast remains challenging, especially in core biopsies. In the current study, we evaluated a spectrum of spindle cell lesion of the breast with a panel of IHC antibodies in an effort to differentiate metaplastic spindle cell carcinoma from its benign and malignant mimickers. Our study included 92 patients who underwent breast core biopsies or breast resections at Northwell Health who were diagnosed with benign and malignant tumor/tumor-like spindle cell lesions. Tumors subtypes in this the study included: angiosarcoma, nodular fasciitis, fibromatosis, myofibroblastoma, phyllodes tumors (benign, borderline and malignant), primary sarcomas and metaplastic spindle cell carcinoma. Our biomarker panel included high molecular weight keratin (HMWK), CAM5.2, AE1/AE3, p63, CD34 and GATA3. GATA3 expression was significantly higher in metaplastic carcinomas (88.9 % vs 4.1 %, p < 0.001), when compared to other spindle cell lesions. The sensitivity and specificity for detecting metaplastic carcinomas reached 84.2 % and 97.3 %, respectively. Regarding cytokeratin panels, none of the three individual markers were as sensitive or specific for metaplastic breast carcinoma. GATA3 is the most specific and sensitive marker forfor the identification of metaplastic spindle cell carcinoma of the breast.
Collapse
Affiliation(s)
- Cao Jin
- Department of Pathology and Laboratory Medicine, Donald and Barbara Zucker School of Medicine at Northwell, United States
| | - Sean Hacking
- Department of Pathology and Laboratory Medicine, Donald and Barbara Zucker School of Medicine at Northwell, United States.
| | - Sujata Sajjan
- Department of Pathology and Laboratory Medicine, Donald and Barbara Zucker School of Medicine at Northwell, United States
| | - Sonia Kamanda
- Department of Pathology and Laboratory Medicine, Donald and Barbara Zucker School of Medicine at Northwell, United States
| | - Tawfiqul Bhuiya
- Department of Pathology and Laboratory Medicine, Donald and Barbara Zucker School of Medicine at Northwell, United States
| | - Mansoor Nasim
- Department of Pathology and Laboratory Medicine, Donald and Barbara Zucker School of Medicine at Northwell, United States
| |
Collapse
|
42
|
Ho NTT, Rahane CS, Pramanik S, Kim PS, Kutzner A, Heese K. FAM72, Glioblastoma Multiforme (GBM) and Beyond. Cancers (Basel) 2021; 13:cancers13051025. [PMID: 33804473 PMCID: PMC7957592 DOI: 10.3390/cancers13051025] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 02/18/2021] [Accepted: 02/22/2021] [Indexed: 12/15/2022] Open
Abstract
Simple Summary Glioblastoma multiforme (GBM) is a serious and aggressive cancer disease that has not allowed scientists to rest for decades. In this review, we consider the new gene pair |-SRGAP2–FAM72-| and discuss its role in the cell cycle and the possibility of defining new therapeutic approaches for the treatment of GBM and other cancers via this gene pair |-SRGAP2–FAM72-|. Abstract Neural stem cells (NSCs) offer great potential for regenerative medicine due to their excellent ability to differentiate into various specialized cell types of the brain. In the central nervous system (CNS), NSC renewal and differentiation are under strict control by the regulation of the pivotal SLIT-ROBO Rho GTPase activating protein 2 (SRGAP2)—Family with sequence similarity 72 (FAM72) master gene (i.e., |-SRGAP2–FAM72-|) via a divergent gene transcription activation mechanism. If the gene transcription control unit (i.e., the intergenic region of the two sub-gene units, SRGAP2 and FAM72) gets out of control, NSCs may transform into cancer stem cells and generate brain tumor cells responsible for brain cancer such as glioblastoma multiforme (GBM). Here, we discuss the surveillance of this |-SRGAP2–FAM72-| master gene and its role in GBM, and also in light of FAM72 for diagnosing various types of cancers outside of the CNS.
Collapse
Affiliation(s)
- Nguyen Thi Thanh Ho
- Graduate School of Biomedical Science and Engineering, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul 133-791, Korea;
| | - Chinmay Satish Rahane
- Maharashtra Institute of Medical Education and Research, Talegaon Dabhade, Maharashtra 410507, India;
| | - Subrata Pramanik
- Institute of Biotechnology, RWTH Aachen University, Worringerweg 3, 52074 Aachen, Germany;
| | - Pok-Son Kim
- Department of Mathematics, Kookmin University, 77 Jeongneung-ro, Seongbuk-gu, Seoul 136-702, Korea;
| | - Arne Kutzner
- Department of Information Systems, College of Computer Science, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul 133-791, Korea;
| | - Klaus Heese
- Graduate School of Biomedical Science and Engineering, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul 133-791, Korea;
- Correspondence:
| |
Collapse
|
43
|
Dong F, Zhao X, Wang J, Huang X, Li X, Zhang L, Dong H, Liu F, Fan M. Dihydroartemisinin inhibits the expression of von Willebrand factor by downregulation of transcription factor ERG in endothelial cells. Fundam Clin Pharmacol 2020; 35:321-330. [PMID: 33107067 PMCID: PMC7983977 DOI: 10.1111/fcp.12622] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Revised: 10/21/2020] [Accepted: 10/22/2020] [Indexed: 12/18/2022]
Abstract
Dihydroartemisinin (DHA), a semi‐synthetic derivative of artemisinin, has effective antitumor and anti‐inflammatory actions. von Willebrand factor (vWF), a large multifunctional glycoprotein, has a prominent function in hemostasis and is a key factor in thrombus formation. In addition, vWF has been regarded as a prospective biomarker for the diagnosis of endothelial dysfunction. In our experiment, we observed that 25 μM DHA specifically downregulated the expression of vWF mRNA and protein in human umbilical vein endothelial cells (HUVECs). Further investigations demonstrated that this DHA‐decreased vWF expression was mediated by the transcription factor ERG and not GATA3. Luciferase activity assay confirmed that DHA regulated the ERG binding with the −56 ETS‐binding motif on the human vWF promoter. Thus, the −56 ETS motif on the vWF promoter region regulates the expression of vWF gene which is induced by DHA. Taken together, we proved that DHA decreased the vWF transcription through the downregulation of ERG in HUVECs. As vWF plays a key role in vascular homeostasis, our findings suggest a new role of DHA in vascular diseases.
Collapse
Affiliation(s)
- Fengyun Dong
- Department of Laboratory Medicine, Affiliated Hospital of Jining Medical University, Jining Medical University, 89 Guhuai Road, Jining, Shandong, 272029, China
| | - Xinghai Zhao
- Shandong Provincial Qianfoshan Hospital, The First Affiliated Hospital of Shandong First Medical University, 16766 Jingshi Road, Jinan, Shandong, 250014, China
| | - Jianning Wang
- Department of Urology, Shandong Provincial Qianfoshan Hospital, The First Affiliated Hospital of Shandong First Medical University, 16766 Jingshi Road, Jinan, Shandong, 250014, China
| | - Xin Huang
- Department of Clinical Pharmacy, Shandong Provincial Qianfoshan Hospital, The First Affiliated Hospital of Shandong First Medical University, 16766 Jingshi Road, Jinan, Shandong, 250014, China
| | - Xiao Li
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, 16369 Jingshi Road, Jinan, Shandong, 250011, China
| | - Liang Zhang
- Shandong Provincial Key Laboratory of Animal Resistance Biology, Institute of Biomedical Sciences, College of Life Sciences, Shandong Normal University, 88 Wenhuadong Street, Jinan, Shandong, 250014, China
| | - Haixin Dong
- Department of Laboratory Medicine, Affiliated Hospital of Jining Medical University, Jining Medical University, 89 Guhuai Road, Jining, Shandong, 272029, China
| | - Fuhong Liu
- Laboratory of Microvascular Medicine, Medical Research Center, Shandong Provincial Qianfoshan Hospital, The First Affiliated Hospital of Shandong First Medical University, 16766 Jingshi Road, Jinan, Shandong, 250014, China
| | - Mengge Fan
- Laboratory of Microvascular Medicine, Medical Research Center, Shandong Provincial Qianfoshan Hospital, The First Affiliated Hospital of Shandong First Medical University, 16766 Jingshi Road, Jinan, Shandong, 250014, China.,Graduate School, Shandong First Medical University & Shandong Academy of Medical Sciences, 6699 Qingdao Road, Jinan, 250000, China
| |
Collapse
|
44
|
Barwe SP, Sidhu I, Kolb EA, Gopalakrishnapillai A. Modeling Transient Abnormal Myelopoiesis Using Induced Pluripotent Stem Cells and CRISPR/Cas9 Technology. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2020; 19:201-209. [PMID: 33102613 PMCID: PMC7558799 DOI: 10.1016/j.omtm.2020.09.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Accepted: 09/13/2020] [Indexed: 01/18/2023]
Abstract
Approximately 1%–2% of children with Down syndrome (DS) develop acute myeloid leukemia (AML) prior to age 5 years. AML in DS children (ML-DS) is characterized by the pathognomonic mutation in the gene encoding the essential hematopoietic transcription factor GATA1, resulting in N-terminally truncated short form of GATA1 (GATA1s). Trisomy 21 and GATA1s together are sufficient to induce transient abnormal myelopoiesis (TAM) exhibiting pre-leukemic characteristics. Approximately 30% of these cases progress into ML-DS by acquisition of additional somatic mutations. We employed disease modeling in vitro by the use of customizable induced pluripotent stem cells (iPSCs) to generate a TAM model. Isogenic iPSC lines derived from the fibroblasts of DS individuals with trisomy 21 and with disomy 21 were used. The CRISPR (Clustered Regularly Interspaced Short Palindromic Repeats)/Cas9 system was used to introduce GATA1 mutation in disomic and trisomic iPSC lines. The hematopoietic stem and progenitor cells (HSPCs) derived from GATA1 mutant iPSC lines expressed GATA1s. The expression of GATA1s concomitant with loss of full-length GATA1 reduced the erythroid population, whereas it augmented megakaryoid and myeloid populations, characteristic of TAM. In conclusion, we have developed a model system representing TAM, which can be used for modeling ML-DS by stepwise introduction of additional mutations.
Collapse
Affiliation(s)
- Sonali P Barwe
- Nemours Center for Childhood Cancer Research, A.I. DuPont Hospital for Children, Wilmington, DE 19803, USA.,University of Delaware, Newark, DE 19711, USA
| | - Ishnoor Sidhu
- Nemours Center for Childhood Cancer Research, A.I. DuPont Hospital for Children, Wilmington, DE 19803, USA.,University of Delaware, Newark, DE 19711, USA
| | - E Anders Kolb
- Nemours Center for Childhood Cancer Research, A.I. DuPont Hospital for Children, Wilmington, DE 19803, USA
| | - Anilkumar Gopalakrishnapillai
- Nemours Center for Childhood Cancer Research, A.I. DuPont Hospital for Children, Wilmington, DE 19803, USA.,University of Delaware, Newark, DE 19711, USA
| |
Collapse
|
45
|
He B, Zhang Y, Zhou Z, Wang B, Liang Y, Lang J, Lin H, Bing P, Yu L, Sun D, Luo H, Yang J, Tian G. A Neural Network Framework for Predicting the Tissue-of-Origin of 15 Common Cancer Types Based on RNA-Seq Data. Front Bioeng Biotechnol 2020; 8:737. [PMID: 32850691 PMCID: PMC7419649 DOI: 10.3389/fbioe.2020.00737] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Accepted: 06/10/2020] [Indexed: 12/19/2022] Open
Abstract
Sequencing-based identification of tumor tissue-of-origin (TOO) is critical for patients with cancer of unknown primary lesions. Even if the TOO of a tumor can be diagnosed by clinicopathological observation, reevaluations by computational methods can help avoid misdiagnosis. In this study, we developed a neural network (NN) framework using the expression of a 150-gene panel to infer the tumor TOO for 15 common solid tumor cancer types, including lung, breast, liver, colorectal, gastroesophageal, ovarian, cervical, endometrial, pancreatic, bladder, head and neck, thyroid, prostate, kidney, and brain cancers. To begin with, we downloaded the RNA-Seq data of 7,460 primary tumor samples across the above mentioned 15 cancer types, with each type of cancer having between 142 and 1,052 samples, from the cancer genome atlas. Then, we performed feature selection by the Pearson correlation method and performed a 150-gene panel analysis; the genes were significantly enriched in the GO:2001242 Regulation of intrinsic apoptotic signaling pathway and the GO:0009755 Hormone-mediated signaling pathway and other similar functions. Next, we developed a novel NN model using the 150 genes to predict tumor TOO for the 15 cancer types. The average prediction sensitivity and precision of the framework are 93.36 and 94.07%, respectively, for the 7,460 tumor samples based on the 10-fold cross-validation; however, the prediction sensitivity and precision for a few specific cancers, like prostate cancer, reached 100%. We also tested the trained model on a 20-sample independent dataset with metastatic tumor, and achieved an 80% accuracy. In summary, we present here a highly accurate method to infer tumor TOO, which has potential clinical implementation.
Collapse
Affiliation(s)
- Binsheng He
- Academician Workstation, Changsha Medical University, Changsha, China
| | | | - Zhen Zhou
- Department of Radiology, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China
| | - Bo Wang
- Geneis (Beijing) Co., Ltd., Beijing, China
| | | | | | - Huixin Lin
- Geneis (Beijing) Co., Ltd., Beijing, China
| | - Pingping Bing
- Academician Workstation, Changsha Medical University, Changsha, China
| | - Lan Yu
- Inner Mongolia People's Hospital, Huhhot, China
| | - Dejun Sun
- Inner Mongolia People's Hospital, Huhhot, China
| | - Huaiqing Luo
- Academician Workstation, Changsha Medical University, Changsha, China
| | - Jialiang Yang
- Academician Workstation, Changsha Medical University, Changsha, China.,Geneis (Beijing) Co., Ltd., Beijing, China
| | - Geng Tian
- Geneis (Beijing) Co., Ltd., Beijing, China
| |
Collapse
|
46
|
Greville G, Llop E, Huang C, Creagh-Flynn J, Pfister S, O'Flaherty R, Madden SF, Peracaula R, Rudd PM, McCann A, Saldova R. Hypoxia Alters Epigenetic and N-Glycosylation Profiles of Ovarian and Breast Cancer Cell Lines in-vitro. Front Oncol 2020; 10:1218. [PMID: 32850359 PMCID: PMC7405916 DOI: 10.3389/fonc.2020.01218] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Accepted: 06/15/2020] [Indexed: 12/14/2022] Open
Abstract
Background: Glycosylation is one of the most fundamental post-translational modifications. Importantly, glycosylation is altered in many cancers. These alterations have been proven to impact on tumor progression and to promote tumor cell survival. From the literature, it is known that there is a clear link between chemoresistance and hypoxia, hypoxia and epigenetics and more recently glycosylation and epigenetics. Methods and Results: Our objective was to investigate these differential parameters, in an in vitro model of ovarian and breast cancer. Ovarian (A2780, A2780cis, PEO1, PEO4) and triple negative breast cancer (TNBC) (MDA-MB-231 and MDA-MB-436) cells were exposed to differential hypoxic conditions (0.5-2% O2) and compared to normoxia (21% O2). Results demonstrated that in hypoxic conditions some significant changes in glycosylation on the secreted N-glycans from the ovarian and breast cancer cell lines were observed. These included, alterations in oligomannosylated, bisected glycans, glycans with polylactosamine extensions, in branching, galactosylation and sialylation in all cell lines except for PEO1. In general, hypoxia exposed ovarian and TNBC cells also displayed increased epithelial to mesenchymal transition (EMT) and migration, with a greater effect seen in the 0.5% hypoxia exposed samples compared to 1 and 2% hypoxia (p ≤ 0.05). SiRNA transient knock down of GATA2/3 transcription factors resulted in a decrease in the expression of glycosyltransferases ST3GAL4 and MGAT5, which are responsible for sialylation and branching, respectively. Conclusions: These glycan changes are known to be integral to cancer cell survival and metastases, suggesting a possible mechanism of action, linking GATA2 and 3, and invasiveness of both ovarian and TNBC cells in vitro.
Collapse
Affiliation(s)
- Gordon Greville
- GlycoScience Group, The National Institute for Bioprocessing Research and Training (NIBRT), Dublin, Ireland.,UCD School of Medicine, College of Health and Agricultural Science (CHAS), University College Dublin (UCD), Dublin, Ireland
| | - Esther Llop
- Biochemistry and Molecular Biology Unit, Department of Biology, University of Girona, Girona, Spain.,Biochemistry of Cancer Group, Girona Biomedical Research Institute (IDIBGI), Girona, Spain
| | - Chengnan Huang
- GlycoScience Group, The National Institute for Bioprocessing Research and Training (NIBRT), Dublin, Ireland
| | - Jack Creagh-Flynn
- UCD School of Medicine, College of Health and Agricultural Science (CHAS), University College Dublin (UCD), Dublin, Ireland
| | - Stephanie Pfister
- UCD School of Medicine, College of Health and Agricultural Science (CHAS), University College Dublin (UCD), Dublin, Ireland
| | - Roisin O'Flaherty
- GlycoScience Group, The National Institute for Bioprocessing Research and Training (NIBRT), Dublin, Ireland
| | - Stephen F Madden
- Data Science Centre, Division of Population Health Sciences, Royal College of Surgeons in Ireland (RCSI), Dublin, Ireland
| | - Rosa Peracaula
- Biochemistry and Molecular Biology Unit, Department of Biology, University of Girona, Girona, Spain.,Biochemistry of Cancer Group, Girona Biomedical Research Institute (IDIBGI), Girona, Spain
| | - Pauline M Rudd
- GlycoScience Group, The National Institute for Bioprocessing Research and Training (NIBRT), Dublin, Ireland.,Analytics Group, Bioprocessing Technology Institute, Astar, Singapore
| | - Amanda McCann
- UCD School of Medicine, College of Health and Agricultural Science (CHAS), University College Dublin (UCD), Dublin, Ireland.,UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin (UCD), Dublin, Ireland
| | - Radka Saldova
- GlycoScience Group, The National Institute for Bioprocessing Research and Training (NIBRT), Dublin, Ireland.,UCD School of Medicine, College of Health and Agricultural Science (CHAS), University College Dublin (UCD), Dublin, Ireland
| |
Collapse
|
47
|
Guo L, Chen B, Zhang G, Wang Y, Cao L, Ren C, Wen L, Lin J, Wei G, Liao N. The transcription factor CBFB mutations indicate an improved survival in HR+/HER2- breast cancer. Gene 2020; 759:144970. [PMID: 32711101 DOI: 10.1016/j.gene.2020.144970] [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] [Received: 04/13/2020] [Revised: 07/09/2020] [Accepted: 07/17/2020] [Indexed: 10/23/2022]
Abstract
BACKGROUND As a critical transcription factor, CBFB (core binding factor subunit β) is frequently mutated in breast cancer and considered to be of significance in the pathogenesis of cancer. The objective of this study was to investigate CBFB mutation profiles and the relationship between CBFB mutations and clinicopathologic characteristics in breast cancer. METHODS A total of 671 treatment-naive Chinese patients with invasive breast cancer at Guangdong Provincial People's Hospital (GDPH) were recruited in this study. CBFB mutation status were detected using the method of capture-based targeted sequencing. Correlation between CBFB mutations and clinicopathologic features were analyzed. Then, we compared the results between Chinese and western population by using Molecular Taxonomy of Breast Cancer International Consortium (METABRIC) cohort (n = 1979) and The Cancer Genome Atlas (TCGA) cohort (n = 925). RESULTS The prevalence of CBFB mutation in GDPH cohort, METABRIC cohort, and TCGA cohort was 4.6% (31/671), 4.6% (92/1979), 2.5% (23/925), respectively. A hotspot mutation due to nucleotide thymine duplication or deletion occurring at the exon2/3 junction was detected in the GDPH and METABRIC cohorts. CBFB mutations were found to be significantly associated with the subtype of HR+/HER2- breast cancer (P = 0.008 in GDPH cohort and P<0.001 in METABRIC cohort), lower tumor grade (P = 0.004 in GDPH cohort and P<0.001 in METABRIC cohort), lower expression of Ki-67 protein (P<0.001 in GDPH cohort), but we didn't find similar results in TCGA cohort. In addition, CBFB in GDPH cohort was observed at a rather high mutation rate in invasive lobular carcinomas (4/18, 22.2%). Further, cox multivariate analysis demonstrated that CBFB was of independent prognosis significance in HR+/HER2- subgroup in METABRIC cohort (HR, 0.562; 95% CI, 0.399-0.790; P = 0.001). CONCLUSION This study reveals race diversity of CBFB mutation spectrum in breast cancers. CBFB mutations mainly occur in HR+/HER2- breast cancer, and it may be a promising prognostic biomarker in HR+/HER2- subgroup.
Collapse
Affiliation(s)
- Liping Guo
- Department of Breast Cancer, Cancer Center, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China; Breast Disease Center, Guangdong Women and Children Hospital, Guangzhou, Guangdong, China
| | - Bo Chen
- Department of Breast Cancer, Cancer Center, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China; School of Medicine, South China University of Technology, Guangzhou, China
| | - Guochun Zhang
- Department of Breast Cancer, Cancer Center, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China; The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China; School of Medicine, South China University of Technology, Guangzhou, China
| | - Yulei Wang
- Department of Breast Cancer, Cancer Center, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Li Cao
- Department of Breast Cancer, Cancer Center, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Chongyang Ren
- Department of Breast Cancer, Cancer Center, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Lingzhu Wen
- Department of Breast Cancer, Cancer Center, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Jiali Lin
- Department of Breast Cancer, Cancer Center, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China; The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Guangnan Wei
- Department of Breast Cancer, Cancer Center, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China; School of Medicine, South China University of Technology, Guangzhou, China
| | - Ning Liao
- Department of Breast Cancer, Cancer Center, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China; The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China; School of Medicine, South China University of Technology, Guangzhou, China.
| |
Collapse
|
48
|
Sakthikumar S, Roy A, Haseeb L, Pettersson ME, Sundström E, Marinescu VD, Lindblad-Toh K, Forsberg-Nilsson K. Whole-genome sequencing of glioblastoma reveals enrichment of non-coding constraint mutations in known and novel genes. Genome Biol 2020; 21:127. [PMID: 32513296 PMCID: PMC7281935 DOI: 10.1186/s13059-020-02035-x] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Accepted: 04/30/2020] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Glioblastoma (GBM) has one of the worst 5-year survival rates of all cancers. While genomic studies of the disease have been performed, alterations in the non-coding regulatory regions of GBM have largely remained unexplored. We apply whole-genome sequencing (WGS) to identify non-coding mutations, with regulatory potential in GBM, under the hypothesis that regions of evolutionary constraint are likely to be functional, and somatic mutations are likely more damaging than in unconstrained regions. RESULTS We validate our GBM cohort, finding similar copy number aberrations and mutated genes based on coding mutations as previous studies. Performing analysis on non-coding constraint mutations and their position relative to nearby genes, we find a significant enrichment of non-coding constraint mutations in the neighborhood of 78 genes that have previously been implicated in GBM. Among them, SEMA3C and DYNC1I1 show the highest frequencies of alterations, with multiple mutations overlapping transcription factor binding sites. We find that a non-coding constraint mutation in the SEMA3C promoter reduces the DNA binding capacity of the region. We also identify 1776 other genes enriched for non-coding constraint mutations with likely regulatory potential, providing additional candidate GBM genes. The mutations in the top four genes, DLX5, DLX6, FOXA1, and ISL1, are distributed over promoters, UTRs, and multiple transcription factor binding sites. CONCLUSIONS These results suggest that non-coding constraint mutations could play an essential role in GBM, underscoring the need to connect non-coding genomic variation to biological function and disease pathology.
Collapse
Affiliation(s)
- Sharadha Sakthikumar
- Department of Medical Biochemistry and Microbiology, Science for Life Laboratory, Uppsala University, SE-751 23, Uppsala, Sweden
- Broad Institute, Cambridge, MA, 02142, USA
| | - Ananya Roy
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, SE-751 85, Uppsala, Sweden
| | - Lulu Haseeb
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, SE-751 85, Uppsala, Sweden
| | - Mats E Pettersson
- Department of Medical Biochemistry and Microbiology, Science for Life Laboratory, Uppsala University, SE-751 23, Uppsala, Sweden
| | - Elisabeth Sundström
- Department of Medical Biochemistry and Microbiology, Science for Life Laboratory, Uppsala University, SE-751 23, Uppsala, Sweden
| | - Voichita D Marinescu
- Department of Medical Biochemistry and Microbiology, Science for Life Laboratory, Uppsala University, SE-751 23, Uppsala, Sweden
| | - Kerstin Lindblad-Toh
- Department of Medical Biochemistry and Microbiology, Science for Life Laboratory, Uppsala University, SE-751 23, Uppsala, Sweden
- Broad Institute, Cambridge, MA, 02142, USA
| | - Karin Forsberg-Nilsson
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, SE-751 85, Uppsala, Sweden.
| |
Collapse
|
49
|
MeCP2 facilitates breast cancer growth via promoting ubiquitination-mediated P53 degradation by inhibiting RPL5/RPL11 transcription. Oncogenesis 2020; 9:56. [PMID: 32483207 PMCID: PMC7264296 DOI: 10.1038/s41389-020-0239-7] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2020] [Revised: 05/13/2020] [Accepted: 05/14/2020] [Indexed: 02/07/2023] Open
Abstract
Methyl-CpG-binding protein 2 (MeCP2) facilitates the carcinogenesis and progression of several types of cancer. However, its role in breast cancer and the relevant molecular mechanism remain largely unclear. In this study, analysis of the Cancer Genome Atlas (TCGA) data that MeCP2 expression was significantly upregulated in breast cancer tissues, and high MeCP2 expression was correlated with poor overall survival. Knockdown of MeCP2 inhibited breast cancer cell proliferation and G1–S cell cycle transition and migration as well as induced cell apoptosis in vitro. Moreover, MeCP2 knockdown suppressed cancer cell growth in vivo. Investigation of the molecular mechanism showed that MeCP2 repressed RPL11 and RPL5 transcription by binding to their promoter regions. TCGA data revealed significantly lower RPL11 and RPL5 expression in breast cancer tissues; additionally, overexpression of RPL11/RPL5 significantly suppressed breast cancer cell proliferation and G1–S cell cycle transition and induced apoptosis in vitro. Furthermore, RPL11 and RPL5 suppressed ubiquitination-mediated P53 degradation through direct binding to MDM2. This study demonstrates that MeCP2 promotes breast cancer cell proliferation and inhibits apoptosis through suppressing RPL11 and RPL5 transcription by binding to their promoter regions.
Collapse
|
50
|
Zhou L, Xu J, Wang S, Yang X, Li C, Zhou J, Zhang P, Xu H, Wang C. Papillary Renal Neoplasm With Reverse Polarity: A Clinicopathologic Study of 7 Cases. Int J Surg Pathol 2020; 28:728-734. [PMID: 32403965 DOI: 10.1177/1066896920918289] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Papillary renal neoplasm with reverse polarity is a form of recently described tumor. These tumors are defined by GATA3 positivity, negative vimentin staining, and the presence of both papillary structures and a layer of eosinophilic cells with apical nuclei and a granular cytoplasm. In the present report, we review 7 cases of papillary renal neoplasm with reverse polarity that were GATA3+ and vimentin-, consistent with past reports. In all 7 of these cases, we found that these tumors were additionally positive for 34βE12. All 7 of these tumors were categorized as stage pT1. On histological examination, these tumors exhibited branching papillae with apical nuclei. All 7 of these patients were alive on most recent follow-up, with 6 being disease free and one having developed prostate cancer. Together, this overview of 7 additional cases of papillary renal neoplasm with reverse polarity offers further insight into this rare and poorly understood disease.
Collapse
Affiliation(s)
- Luting Zhou
- Ruijin Hospital, Shanghai Jiaotong University, Shanghai, China
| | - Jiankun Xu
- Ruijin Hospital, Shanghai Jiaotong University, Shanghai, China
| | - Suying Wang
- Ningbo Diagnostic Pathology Center, Zhejiang Province, Ningbo, China
| | - Xiaoqun Yang
- Ruijin Hospital, Shanghai Jiaotong University, Shanghai, China
| | - Chuanying Li
- Ruijin Hospital, Shanghai Jiaotong University, Shanghai, China
| | - Jun Zhou
- Ruijin Hospital, Shanghai Jiaotong University, Shanghai, China
| | - Peipei Zhang
- Ruijin Hospital, Shanghai Jiaotong University, Shanghai, China
| | - Haimin Xu
- Ruijin Hospital, Shanghai Jiaotong University, Shanghai, China
| | - Chaofu Wang
- Ruijin Hospital, Shanghai Jiaotong University, Shanghai, China
| |
Collapse
|