1
|
Bailey-Lundberg JM. Decoding Ribosome Stress: How NAT10 Influences Pancreatic Acinar Cell Survival and Cancer Development. Cell Mol Gastroenterol Hepatol 2025:101516. [PMID: 40318699 DOI: 10.1016/j.jcmgh.2025.101516] [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: 03/28/2025] [Accepted: 04/08/2025] [Indexed: 05/07/2025]
Affiliation(s)
- Jennifer M Bailey-Lundberg
- Department of Pathology, Microbiology and Immunology, The University of Nebraska Medical Center, Omaha, Nebraska; Fred and Pamela Buffett Cancer Center, The University of Nebraska Medical Center, Omaha, Nebraska.
| |
Collapse
|
2
|
Li J, Dong J, Li M, Zhu H, Xin P. Potential mechanisms for predicting comorbidity between multiple myeloma and femoral head necrosis based on multiple bioinformatics. Comput Biol Chem 2024; 113:108220. [PMID: 39405776 DOI: 10.1016/j.compbiolchem.2024.108220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Revised: 09/07/2024] [Accepted: 09/18/2024] [Indexed: 12/15/2024]
Abstract
OBJECTIVE This study aims to utilize multiple bioinformatics tools to elucidate the potential mechanisms underlying the comorbidity of Multiple Myeloma (MM) and Osteonecrosis of the Femoral Head (ONFH). METHOD High-throughput microarray datasets for MM and ONFH were retrieved from the GEO database, followed by separate preprocessing. We applied Weighted Gene Co-expression Network Analysis (WGCNA) to construct co-expression networks within the MM datasets, further identifying modules and genes associated with MM clinical characteristics. Potential comorbid genes were enriched and analyzed using pathway and network analysis tools, and key genes for MM and ONFH comorbidity were preliminarily screened using Cytoscape. The gene expression capabilities and performance were validated using two disease-related datasets, and we evaluated the differences and consistencies in the immune microenvironment between the two diseases. RESULTS Our screening identified 418 immune-related comorbid genes, showing consistent biological processes in ribosome synthesis, particularly protein synthesis across both diseases. Key genes were further identified through Protein-Protein Interaction (PPI) networks, and their performance was validated in a validation cohort, with Receiver Operating Characteristic (ROC) curve areas exceeding 0.8. The immune microenvironment analysis highlighted consistent plasma cell infiltration correlated with disease comorbidity, suggesting potential immune targets for future therapies. CONCLUSION MM and ONFH share common pathogenic genes that mediate changes in signaling pathways and immune cell dynamics, potentially influencing the comorbidity and progression of these diseases. Key genes identified, such as RPS19, RPL35, RPL24, RPL36, and EIF3G, along with plasma cell infiltration, may serve as central mechanisms in the development of both diseases. This study offers insights and references for further research into targeted treatments for these conditions.
Collapse
Affiliation(s)
- Jie Li
- The Second Affiliated Hospital of Shandong First Medical University(Department of Hematology), Tai'an, Shandong 271000, China
| | - Jing Dong
- The Second Affiliated Hospital of Shandong First Medical University(Department of Hematology), Tai'an, Shandong 271000, China
| | - Ming Li
- The Second Affiliated Hospital of Shandong First Medical University(Department of Orthopedics), Tai'an, Shandong 271000, China
| | - Hongbo Zhu
- The Second Affiliated Hospital of Shandong First Medical University(Department of Hematology), Tai'an, Shandong 271000, China
| | - Peicheng Xin
- The Second Affiliated Hospital of Shandong First Medical University(Department of Orthopedics), Tai'an, Shandong 271000, China.
| |
Collapse
|
3
|
Patel P, Nandi A, Verma SK, Kaushik N, Suar M, Choi EH, Kaushik NK. Zebrafish-based platform for emerging bio-contaminants and virus inactivation research. THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 872:162197. [PMID: 36781138 PMCID: PMC9922160 DOI: 10.1016/j.scitotenv.2023.162197] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 01/23/2023] [Accepted: 02/08/2023] [Indexed: 05/27/2023]
Abstract
Emerging bio-contaminants such as viruses have affected health and environment settings of every country. Viruses are the minuscule entities resulting in severe contagious diseases like SARS, MERS, Ebola, and avian influenza. Recent epidemic like the SARS-CoV-2, the virus has undergone mutations strengthen them and allowing to escape from the remedies. Comprehensive knowledge of viruses is essential for the development of targeted therapeutic and vaccination treatments. Animal models mimicking human biology like non-human primates, rats, mice, and rabbits offer competitive advantage to assess risk of viral infections, chemical toxins, nanoparticles, and microbes. However, their economic maintenance has always been an issue. Furthermore, the redundancy of experimental results due to aforementioned aspects is also in examine. Hence, exploration for the alternative animal models is crucial for risk assessments. The current review examines zebrafish traits and explores the possibilities to monitor emerging bio-contaminants. Additionally, a comprehensive picture of the bio contaminant and virus particle invasion and abatement mechanisms in zebrafish and human cells is presented. Moreover, a zebrafish model to investigate the emerging viruses such as coronaviridae and poxviridae has been suggested.
Collapse
Affiliation(s)
- Paritosh Patel
- Plasma Bioscience Research Center, Department of Electrical and Biological Physics, Kwangwoon University, 01897 Seoul, South Korea
| | - Aditya Nandi
- School of Biotechnology, KIIT University, Bhubaneswar 751024, Odisha, India
| | - Suresh K Verma
- School of Biotechnology, KIIT University, Bhubaneswar 751024, Odisha, India; Condensed Matter Theory Group, Materials Theory Division, Department of Physics and Astronomy, Uppsala University, Box 516, SE-751 20 Uppsala, Sweden
| | - Neha Kaushik
- Department of Biotechnology, College of Engineering, The University of Suwon, 18323 Hwaseong, Republic of Korea
| | - Mrutyunjay Suar
- School of Biotechnology, KIIT University, Bhubaneswar 751024, Odisha, India
| | - Eun Ha Choi
- Plasma Bioscience Research Center, Department of Electrical and Biological Physics, Kwangwoon University, 01897 Seoul, South Korea.
| | - Nagendra Kumar Kaushik
- Plasma Bioscience Research Center, Department of Electrical and Biological Physics, Kwangwoon University, 01897 Seoul, South Korea.
| |
Collapse
|
4
|
Ghufran M, Khan HA, Ullah M, Ghufran S, Ayaz M, Siddiq M, Hassan SSU, Bungau S. In Silico Strategies for Designing of Peptide Inhibitors of Oncogenic K-Ras G12V Mutant: Inhibiting Cancer Growth and Proliferation. Cancers (Basel) 2022; 14:4884. [PMID: 36230807 PMCID: PMC9564332 DOI: 10.3390/cancers14194884] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 10/02/2022] [Accepted: 10/03/2022] [Indexed: 12/12/2022] Open
Abstract
Ras plays a pivotal function in cell proliferation and is an important protein in signal transduction pathways. Mutations in genes encoding the Ras protein drive the signaling cascades essential for malignant transformation, tumour angiogenesis, and metastasis and are responsible for above 30% of all human cancers. There is evidence that N-Ras, K-Ras, and H-Ras play significant roles in human cancer. The mutated K-Ras protein is typically observed in malignant growths. Mutant K-Ras is the most common in lung, colon, and pancreatic cancers. The purpose of this research was to create peptides that inhibit K-Ras G12V. The crystal structure of the mutant K-Ras G12V-H-REV107 complex was obtained from a protein data bank. Further, we used a residue scan approach to create unique peptides from the reference peptide (H-REV107). AMBER molecular dynamics simulations were used to test the stability of the top four proposed peptides (based on binding free energies). Our findings showed that the top four selected peptides had stronger interactions with K-Ras than the reference peptide and have the ability to block the activation function of K-Ras. Our extensive analyses of binding affinities showed that our designed peptide possesses the potential to inhibit K-Ras and to reduce the progression of cancer.
Collapse
Affiliation(s)
- Mehreen Ghufran
- Department of Pathology, Medical Teaching Institution Bacha Khan Medical College (BKMC) Mardan, Mardan 23200, Pakistan
| | - Haider Ali Khan
- Department of Biochemistry, Abdul Wali Khan University Mardan, Mardan 23200, Pakistan
| | - Mehran Ullah
- District Medical Officer, Sehat Sahulat Program (SSP), KPK, Mardan 23200, Pakistan
- District Headquarter (DHQ) Hospital Mardan, Mardan 23200, Pakistan
| | - Sabreen Ghufran
- Department of Biochemistry, Abdul Wali Khan University Mardan, Mardan 23200, Pakistan
| | - Muhammad Ayaz
- Department of Pharmacy, Faculty of Biological Sciences, University of Malakand, Chakdara 18800, Khyber Pakhtunkhwa, Pakistan
| | - Muhammad Siddiq
- Department of Pharmacy, Abdul Wali Khan University Mardan, Mardan 23200, Pakistan
| | - Syed Shams ul Hassan
- Shanghai Key Laboratory for Molecular Engineering of Chiral Drugs, School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China
- Department of Natural Product Chemistry, School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Simona Bungau
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, 410028 Oradea, Romania
| |
Collapse
|
5
|
Jia D, Zhang R, Shao J, Zhang W, Cai L, Sun W. Exposure to trace levels of metals and fluoroquinolones increases inflammation and tumorigenesis risk of zebrafish embryos. ENVIRONMENTAL SCIENCE AND ECOTECHNOLOGY 2022; 10:100162. [PMID: 36159734 PMCID: PMC9488011 DOI: 10.1016/j.ese.2022.100162] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 02/17/2022] [Accepted: 02/18/2022] [Indexed: 05/04/2023]
Abstract
Exposure to trace-level heavy metals and antibiotics may elicit metabolic disorder, alter protein expression, and then induce pathological changes in zebrafish embryos, despite negligible physiological and developmental toxicity. This study investigated the single and combined developmental toxicity of fluoroquinolones (enrofloxacin [ENR] and ciprofloxacin [CIP]) (≤0.5 μM) and heavy metals (Cu and Cd) (≤0.5 μM) to zebrafish embryos, and molecular responses of zebrafish larvae upon exposure to the single pollutant (0.2 μM) or a binary metal-fluoroquinolone mixture (0.2 μM). In all single and mixture exposure groups, no developmental toxicity was observed, but oxidative stress, inflammation, and lipid depletion were found in zebrafish embryos, which was more severe in the mixture exposure groups than in the single exposure groups, probably due to increased metal bioaccumulation in the presence of ENR or CIP. Metabolomics analysis revealed the up-regulation of amino acids and down-regulation of fatty acids, corresponding to an active response to oxidative stress and the occurrence of inflammation. The up-regulation of antioxidase and immune proteins revealed by proteomics analysis further confirmed the occurrence of oxidative stress and inflammation. Furthermore, the KEGG pathway enrichment analysis showed a significant disturbance of pathways related to immunity and tumor, indicating the potential risk of tumorigenesis in zebrafish larvae. The findings provide molecular-level insights into the adverse effects of heavy metals and antibiotics (especially in chemical mixtures) on zebrafish embryos, and highlight the potential ecotoxicological risks of trace-level heavy metals and antibiotics in the environment.
Collapse
Affiliation(s)
- Dantong Jia
- College of Environmental Sciences and Engineering, Peking University, The Key Laboratory of Water and Sediment Sciences, Ministry of Education, Beijing, 100871, China
- State Environmental Protection Key Laboratory of All Material Fluxes in River Ecosystems, International Joint Laboratory for Regional Pollution Control, Ministry of Education, Beijing, 100871, China
| | - Ruijie Zhang
- College of Environmental Sciences and Engineering, Peking University, The Key Laboratory of Water and Sediment Sciences, Ministry of Education, Beijing, 100871, China
- State Environmental Protection Key Laboratory of All Material Fluxes in River Ecosystems, International Joint Laboratory for Regional Pollution Control, Ministry of Education, Beijing, 100871, China
| | - Jian Shao
- College of Animal Science, Guizhou University, The Key Laboratory of Animal Genetics, Breeding and Reproduction in the Plateau Mountainous Region, Ministry of Education, Guiyang, Guizhou, 550025, China
| | - Wei Zhang
- Department of Plant, Soil and Microbial Sciences, Environmental Science and Policy Program, Michigan State University, East Lansing, MI, 48824, United States
| | - Leilei Cai
- College of Safety and Environmental Engineering, Shandong University of Science and Technology, Qingdao, Shandong, 266590, China
| | - Weiling Sun
- College of Environmental Sciences and Engineering, Peking University, The Key Laboratory of Water and Sediment Sciences, Ministry of Education, Beijing, 100871, China
- State Environmental Protection Key Laboratory of All Material Fluxes in River Ecosystems, International Joint Laboratory for Regional Pollution Control, Ministry of Education, Beijing, 100871, China
- Corresponding author. Peking University. China.
| |
Collapse
|
6
|
Liu J, Xiao S, Chen J. Development of an Inflammation-Related lncRNA-miRNA-mRNA Network Based on Competing Endogenous RNA in Breast Cancer at Single-Cell Resolution. Front Cell Dev Biol 2022; 10:839876. [PMID: 35145966 PMCID: PMC8821924 DOI: 10.3389/fcell.2022.839876] [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: 12/20/2021] [Accepted: 01/06/2022] [Indexed: 12/14/2022] Open
Abstract
The role and mechanism of inflammation in breast cancer is unclear. This study aims to probe the relationship between inflammation and long non-coding RNAs (lncRNAs) and to stablish an inflammation-related competing endogenous RNA (ceRNA) network in breast cancer. Inflammation-related lncRNAs and target genes were screened based on the data from four single-cell RNA sequencing (scRNA-seq) studies and miRNAs were bioinformatically predicted according to ceRNA hypothesis. A series of in silico analyses were performed to construct an inflammation-related ceRNA network in breast cancer. Consequently, a total of seven inflammation-related lncRNAs were selected, after which LRRC75A-AS1 was identified as the most potential lncRNA in view of its expression and prognostic predictive value in breast cancer. Finally, an inflammation-related ceRNA network in breast cancer at the single cell level was established based on lncRNA LRRC75A-AS1, miR-3127-5p, miR-2114-3p, RPL36 and RPL27A mRNAs. Collectively, the lncRNA LRRC75A-AS1 and the LRRC75A-AS1-based on ceRNA network may exert crucial roles in modulating inflammation response during the initiation and progression of breast cancer.
Collapse
Affiliation(s)
- Jingxing Liu
- Department of Intensive Care Unit, Changxing People's Hospital of Zhejiang, Huzhou, China
| | - Shuyuan Xiao
- Department of Anesthesiology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Jing Chen
- Department of Oncology, The First Affiliated Hospital of Jiaxing University, Jiaxing, China
| |
Collapse
|
7
|
The Identification of Zinc-Finger Protein 433 as a Possible Prognostic Biomarker for Clear-Cell Renal Cell Carcinoma. Biomolecules 2021; 11:biom11081193. [PMID: 34439859 PMCID: PMC8392881 DOI: 10.3390/biom11081193] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 07/31/2021] [Accepted: 08/06/2021] [Indexed: 11/18/2022] Open
Abstract
Clear-cell renal cell carcinoma (ccRCC) is the most common and aggressive form of all urological cancers, with poor prognosis and high mortality. At late stages, ccRCC is known to be mainly resistant to chemotherapy and radiotherapy. Therefore, it is urgent and necessary to identify biomarkers that can facilitate the early detection of ccRCC in patients. In this study, the levels of transcripts of ccRCC from The Cancer Genome Atlas (TCGA) dataset were used to identify prognostic biomarkers in this disease. Analyzing the data obtained indicated that the KRAB-ZNF protein is significantly suppressed in clear-cell carcinomas. Furthermore, ZNF433 is differentially expressed in ccRCC in a stage- and histological-grade-specific manner. In addition, ZNF433 expression was correlated with metastasis, with greater node involvement associated with lower ZNF433 expression (p < 0.01) and with a more unsatisfactory overall survival outcome (HR, 0.45; 95% CI, 0.33–0.6; p = 8.5 × 10−8). Since ccRCC is characterized by mutations in proteins that alter epigenetic modifications and /or chromatin remodeling, we examined the expression of ZNF433 transcripts in ccRCC with wildtype and mutated forms of BAP1, KDMC5, MTOR, PBRM1, SETD2, and VHL. Analysis revealed that ZNF433 expression was significantly reduced in ccRCC with mutations in the BAP1, SETD2, and KDM5C genes (p < 0.05). In addition, the ZNF433 promoter region was highly methylated, and hypermethylation was significantly associated with mRNA suppression (p < 2.2 × 10−16). In silico analysis of potential ZNF target genes found that the largest group of target genes are involved in cellular metabolic processes, which incidentally are particularly impaired in ccRCC. It was concluded from this study that gene expression of ZNF433 is associated with cancer progression and poorer prognosis, and that ZNF433 behaves in a manner that suggests that it is a prognostic marker and a possible tumor-suppressor gene in clear-cell renal cell carcinoma.
Collapse
|
8
|
Fang Y, Zong Q, He Z, Liu C, Wang YF. Knockdown of RpL36 in testes impairs spermatogenesis in Drosophila melanogaster. JOURNAL OF EXPERIMENTAL ZOOLOGY PART B-MOLECULAR AND DEVELOPMENTAL EVOLUTION 2021; 336:417-430. [PMID: 33734578 DOI: 10.1002/jez.b.23040] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2019] [Revised: 02/06/2021] [Accepted: 02/27/2021] [Indexed: 01/03/2023]
Abstract
Many ribosomal proteins (RPs) not only play essential roles in ribosome biogenesis, but also have "extraribosomal" functions in various cellular processes. RpL36 encodes ribosomal protein L36, a component of the 60S subunit of ribosomes in Drosophila melanogaster. We report here that RpL36 is required for spermatogenesis in D. melanogaster. After showing the evolutionary conservation of RpL36 sequences in animals, we revealed that the RpL36 expression level in fly testes was significantly higher than in ovaries. Knockdown RpL36 in fly testes resulted in a significantly decreased egg hatch rate when these males mated with wild-type females. Furthermore, 76.67% of the RpL36 knockdown fly testes were much smaller in comparison to controls. Immunofluorescence staining exhibited that in the RpL36 knockdown testis hub cell cluster was enlarged, while the number of germ cells, including germ stem cells, was reduced. Knockdown of RpL36 in fly testis caused much fewer or no mature sperms in seminal vesicles. The terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) signal was stronger in RpL36 knockdown fly testes than in the control testes, but the TUNEL-positive cells could not be stained by Vasa antibody, indicating that apoptotic cells are not germ cells. The percentage of pH3-positive cells among the Vasa-positive cells was significantly reduced. The expression of genes involved in cell death, cell cycle progression, and JAK/STAT signaling pathway was significantly changed by RpL36 knockdown in fly testes. These results suggest that RpL36 plays an important role in spermatogenesis, likely through JAK/STAT pathway, thus resulting in defects in cell-cycle progression and cell death in D. melanogaster testes.
Collapse
Affiliation(s)
- Yang Fang
- School of Life Sciences, Hubei Key Laboratory of Genetic Regulation and Integrative Biology, Central China Normal University, Wuhan, China
| | - Qiong Zong
- School of Life Sciences, Hubei Key Laboratory of Genetic Regulation and Integrative Biology, Central China Normal University, Wuhan, China
| | - Zhen He
- School of Life Sciences, Hubei Key Laboratory of Genetic Regulation and Integrative Biology, Central China Normal University, Wuhan, China
| | - Chen Liu
- Institute of Biology and Medicine, Wuhan University of Science and Technology, Wuhan, China
| | - Yu-Feng Wang
- School of Life Sciences, Hubei Key Laboratory of Genetic Regulation and Integrative Biology, Central China Normal University, Wuhan, China
| |
Collapse
|
9
|
Abstract
Zebrafish are rapidly becoming a leading model organism for cancer research. The genetic pathways driving cancer are highly conserved between zebrafish and humans, and the ability to easily manipulate the zebrafish genome to rapidly generate transgenic animals makes zebrafish an excellent model organism. Transgenic zebrafish containing complex, patient-relevant genotypes have been used to model many cancer types. Here we present a comprehensive review of transgenic zebrafish cancer models as a resource to the field and highlight important areas of cancer biology that have yet to be studied in the fish. The ability to image cancer cells and niche biology in an endogenous tumor makes zebrafish an indispensable model organism in which we can further understand the mechanisms that drive tumorigenesis and screen for potential new cancer therapies.
Collapse
Affiliation(s)
- Alicia M. McConnell
- Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital and Dana-Farber Cancer Institute, Boston, Massachusetts 02115, USA
- Harvard Stem Cell Institute, Boston, Massachusetts 02138, USA
- Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Haley R. Noonan
- Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital and Dana-Farber Cancer Institute, Boston, Massachusetts 02115, USA
- Harvard Stem Cell Institute, Boston, Massachusetts 02138, USA
- Harvard Medical School, Boston, Massachusetts 02115, USA
- Biological and Biomedical Sciences Program, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Leonard I. Zon
- Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital and Dana-Farber Cancer Institute, Boston, Massachusetts 02115, USA
- Harvard Stem Cell Institute, Boston, Massachusetts 02138, USA
- Harvard Medical School, Boston, Massachusetts 02115, USA
- Stem Cell and Regenerative Biology Department and Howard Hughes Medical Institute, Harvard University, Boston, Massachusetts 02138, USA
| |
Collapse
|
10
|
Lessard F, Brakier-Gingras L, Ferbeyre G. Ribosomal Proteins Control Tumor Suppressor Pathways in Response to Nucleolar Stress. Bioessays 2019; 41:e1800183. [PMID: 30706966 DOI: 10.1002/bies.201800183] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 12/18/2018] [Indexed: 01/05/2023]
Abstract
Ribosome biogenesis includes the making and processing of ribosomal RNAs, the biosynthesis of ribosomal proteins from their mRNAs in the cytosol and their transport to the nucleolus to assemble pre-ribosomal particles. Several stresses including cellular senescence reduce nucleolar rRNA synthesis and maturation increasing the availability of ribosome-free ribosomal proteins. Several ribosomal proteins can activate the p53 tumor suppressor pathway but cells without p53 can still arrest their proliferation in response to an imbalance between ribosomal proteins and mature rRNA production. Recent results on senescence-associated ribogenesis defects (SARD) show that the ribosomal protein S14 (RPS14 or uS11) can act as a CDK4/6 inhibitor linking ribosome biogenesis defects to the main engine of cell cycle progression. This work offers new insights into the regulation of the cell cycle and suggests novel avenues to design anticancer drugs.
Collapse
Affiliation(s)
- Frédéric Lessard
- Department of Biochemistry and Molecular Medicine, Université de Montréal, C.P. 6128, Succ. Centre-Ville, Montréal, Québec H3C 3J7, Canada
| | - Léa Brakier-Gingras
- Department of Biochemistry and Molecular Medicine, Université de Montréal, C.P. 6128, Succ. Centre-Ville, Montréal, Québec H3C 3J7, Canada
| | - Gerardo Ferbeyre
- Department of Biochemistry and Molecular Medicine, Université de Montréal, C.P. 6128, Succ. Centre-Ville, Montréal, Québec H3C 3J7, Canada.,CRCHUM, 900 Saint-Denis - bureau R10.432, Montréal, Québec H2X 0A9, Canada
| |
Collapse
|
11
|
Fu XH, Chen ZT, Wang WH, Fan XJ, Huang Y, Wu XB, Huang JL, Wang JX, Lin HJ, Tan XL, Wang L, Wang JP. KRAS G12V Mutation is an Adverse Prognostic Factor of Chinese Gastric Cancer Patients. J Cancer 2019; 10:821-828. [PMID: 30854087 PMCID: PMC6400811 DOI: 10.7150/jca.27899] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Accepted: 12/14/2018] [Indexed: 12/13/2022] Open
Abstract
This study aims to investigate the molecular characteristics of Chinese gastric cancer patients. In our study, the KRAS, BRAF, and PIK3CA mutation status of 485 GC patients were analyzed by Sanger sequencing. Kaplan-Meier analysis was used to plot survival curves according to different genotypes. The results show that the frequency of KRAS, BRAF and PIK3CA mutations were 4.1%, 1.2% and 3.5%, respectively. BRAF mutations were significantly concentrated in stage III and IV gastric cancer (P=0.009). KRAS G12V mutation carriers have much shorter OS than other mutation carriers and wild-type group patients (P=0.013). In conclusion, only the KRAS G12V mutation has an adverse effect on patient survival.
Collapse
Affiliation(s)
- Xin-Hui Fu
- Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangdong, China.,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangdong, China.,Department of Pathology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangdong, China
| | - Zhi-Ting Chen
- Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangdong, China.,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangdong, China.,Department of Pathology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangdong, China
| | - Wen-Hui Wang
- Department of Information and Technology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangdong, China
| | - Xin-Juan Fan
- Department of Pathology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangdong, China
| | - Yan Huang
- Department of Pathology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangdong, China
| | - Xiao-Bin Wu
- Department of GI Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangdong, China
| | - Jing-Lin Huang
- Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangdong, China.,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangdong, China.,Department of Pathology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangdong, China
| | - Jing-Xuan Wang
- Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangdong, China.,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangdong, China.,Department of Pathology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangdong, China
| | - Han-Jie Lin
- Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangdong, China.,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangdong, China.,Department of Pathology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangdong, China
| | - Xiao-Li Tan
- Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangdong, China.,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangdong, China.,Department of Pathology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangdong, China
| | - Lei Wang
- Department of GI Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangdong, China
| | - Jian-Ping Wang
- Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangdong, China.,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangdong, China
| |
Collapse
|
12
|
Zhou Y, Tong L, Wang M, Chang X, Wang S, Li K, Xiao J. miR-505-3p is a repressor of the puberty onset in female mice. J Endocrinol 2018; 240:JOE-18-0533.R2. [PMID: 30557853 DOI: 10.1530/joe-18-0533] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Accepted: 12/17/2018] [Indexed: 12/31/2022]
Abstract
Puberty onset is a complex trait regulated by multiple genetic and environmental factors. In this study, we narrowed a puberty related QTL region down to a 1.7 Mb region on chromosome X in female mice and inferred miR-505-3p as the functional gene. We conducted ectopic expression of miR-505-3p in the hypothalamus of prepubertal female mice through lentivirus-mediated orthotopic injection. The impact of miR-505-3p on female puberty was evaluated by the measurement of pubertal/reproduction events and histological analysis. The results showed that female mice with overexpression of miR-505-3p in the hypothalamus manifested later puberty onset timing both in vaginal opening and ovary maturation, followed by weaker fertility lying in the longer interval time between mating and delivery, higher abortion rate and smaller litter size. We also constructed miR-505-3p knockout mice by CRISPR/Cas9 technology. MiR-505-3p knockout female mice showed earlier vaginal opening timing, higher serum gonadotrophin and higher expression of puberty-related gene in the hypothalamus than their wild type littermates. Srsf1 was proved to be the target gene of miR-505-3p that played the major role in this process. The results of RNA Immunoprecipitation-sequencing showed that SRSF1 (or SF2), the protein product of Srsf1 gene, mainly bound to ribosome protein (RP) mRNAs in GT1-7 cells. The collective evidence implied that miR-505-3p/SRSF1/RP could play a role in the sexual maturation regulation of mammals.
Collapse
Affiliation(s)
- Yuxun Zhou
- Y Zhou, The College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai, China
| | - Li Tong
- L Tong, The College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai, China
| | - Maochun Wang
- M Wang, The College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai, China
| | - Xueying Chang
- X Chang, The College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai, China
| | - Sijia Wang
- S Wang, The College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai, China
| | - Kai Li
- K Li, Department of Bioengineer, Donghua University, Songjiang, 201620, China
| | - Junhua Xiao
- J Xiao, The College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai, China
| |
Collapse
|
13
|
Lu JW, Raghuram D, Fong PSA, Gong Z. Inducible Intestine-Specific Expression of kras V12 Triggers Intestinal Tumorigenesis In Transgenic Zebrafish. Neoplasia 2018; 20:1187-1197. [PMID: 30390498 PMCID: PMC6215966 DOI: 10.1016/j.neo.2018.10.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2018] [Revised: 10/10/2018] [Accepted: 10/11/2018] [Indexed: 02/06/2023] Open
Abstract
KRAS mutations are a major risk factor in colorectal cancers. In particular, a point mutation of KRAS of amino acid 12, such as KRASV12, renders it stable activity in oncogenesis. We found that krasV12 promotes intestinal carcinogenesis by generating a transgenic zebrafish line with inducible krasV12 expression in the intestine, Tg(ifabp:EGFP-krasV12). The transgenic fish generated exhibited significant increases in the rates of intestinal epithelial outgrowth, proliferation, and cross talk in the active Ras signaling pathway involving in epithelial-mesenchymal transition (EMT). These results provide in vivo evidence of Ras pathway activation via krasV12 overexpression. Long-term transgenic expression of krasV12 resulted in enteritis, epithelial hyperplasia, and tubular adenoma in adult fish. This was accompanied by increased levels of the signaling proteins p-Erk and p-Akt and by downregulation of the EMT marker E-cadherin. Furthermore, we also observed a synergistic effect of krasV12 expression and dextran sodium sulfate treatment to enhance intestinal tumor in zebrafish. Our results demonstrate that krasV12 overexpression induces intestinal tumorigenesis in zebrafish, which mimics intestinal tumor formation in humans. Thus, our transgenic zebrafish may provide a valuable in vivo platform that can be used to investigate tumor initiation and anticancer drugs for gastrointestinal cancers.
Collapse
Affiliation(s)
- Jeng-Wei Lu
- Department of Biological Sciences, National University of Singapore, Singapore
| | - Divya Raghuram
- Department of Biological Sciences, National University of Singapore, Singapore
| | | | - Zhiyuan Gong
- Department of Biological Sciences, National University of Singapore, Singapore.
| |
Collapse
|
14
|
Hu Y, Kang C, Zhao J, Nie Y, Zheng L, Li H, Li X, Wang Q, Qiu Y. LncRNA PLAC2 down-regulates RPL36 expression and blocks cell cycle progression in glioma through a mechanism involving STAT1. J Cell Mol Med 2018; 22:497-510. [PMID: 28922548 PMCID: PMC5742712 DOI: 10.1111/jcmm.13338] [Citation(s) in RCA: 85] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Accepted: 07/05/2017] [Indexed: 12/20/2022] Open
Abstract
Current glioma therapies allow in situ delivery of cytotoxic drugs to the tumour; however, gliomas show early recurrence due to their highly proliferative character. Long non-coding (lnc)RNAs play critical roles in tumorigenesis by controlling cell proliferation and cycling. However, the mechanism of action of lncRNAs in glioma development remains unclear. Here, we report that the lncRNA PLAC2 induces cell cycle arrest by targeting ribosomal protein (RP)L36 in glioma. RPL36 promoted cell proliferation and G1/S cell cycle progression. Mass spectrometry analysis revealed that signal transducer and activator of transcription (STAT)1 interacted with both lncRNA PLAC2 and the RPL36 promoter. We also found that the nucleus PLAC2 bind with STAT1 and interact with RPL36 promoters but the cytoplasmic lncRNA PLAC2 inhibited STAT1 nuclear transfer, thereby decreasing RP36 expression, inhibiting cell proliferation and inducing cell cycle arrest. These results provide evidence for a novel cell cycle regulatory network in glioma comprising the lncRNA PLAC2 along with STAT1 and RPL36 that can serve as a therapeutic target for glioma treatment.
Collapse
Affiliation(s)
- Yan‐Wei Hu
- Laboratory Medicine CenterNanfang HospitalSouthern Medical UniversityGuangzhouGuangdongChina
| | - Chun‐Min Kang
- Laboratory Medicine CenterNanfang HospitalSouthern Medical UniversityGuangzhouGuangdongChina
| | - Jing‐Jing Zhao
- Laboratory Medicine CenterNanfang HospitalSouthern Medical UniversityGuangzhouGuangdongChina
| | - Ying Nie
- Department of AnesthesiologyGuangdong 999 Brain HospitalGuangzhouGuangdongChina
| | - Lei Zheng
- Laboratory Medicine CenterNanfang HospitalSouthern Medical UniversityGuangzhouGuangdongChina
| | - Hai‐Xia Li
- Laboratory Medicine CenterNanfang HospitalSouthern Medical UniversityGuangzhouGuangdongChina
| | - Xin Li
- Laboratory Medicine CenterNanfang HospitalSouthern Medical UniversityGuangzhouGuangdongChina
| | - Qian Wang
- Laboratory Medicine CenterNanfang HospitalSouthern Medical UniversityGuangzhouGuangdongChina
| | - Yu‐Rong Qiu
- Laboratory Medicine CenterNanfang HospitalSouthern Medical UniversityGuangzhouGuangdongChina
| |
Collapse
|
15
|
Astone M, Dankert EN, Alam SK, Hoeppner LH. Fishing for cures: The alLURE of using zebrafish to develop precision oncology therapies. NPJ Precis Oncol 2017; 1. [PMID: 29376139 PMCID: PMC5784449 DOI: 10.1038/s41698-017-0043-9] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Zebrafish have proven to be a valuable model to study human cancer biology with the ultimate aim of developing new therapies. Danio rerio are amenable to in vivo imaging, high-throughput drug screening, mutagenesis, and transgenesis, and they share histological and genetic similarities with Homo sapiens. The significance of zebrafish in the field of precision oncology is rapidly emerging. Indeed, modeling cancer in zebrafish has already been used to identify tumor biomarkers, define therapeutic targets and provide an in vivo platform for drug discovery. New zebrafish studies are starting to pave the way to direct individualized clinical applications. Patient-derived cancer cell xenograft models have demonstrated the feasibility of using zebrafish as a real-time avatar of prognosis and drug response to identify the most ideal therapy for an individual patient. Genetic cancer modeling in zebrafish, now facilitated by rapidly evolving genome editing techniques, represents another innovative approach to recapitulate human oncogenesis and develop individualized treatments. Utilizing zebrafish to design customizable precision therapies will improve the clinical outcome of patients afflicted with cancer.
Collapse
Affiliation(s)
- Matteo Astone
- The Hormel Institute, University of Minnesota, Austin, MN, 55912
| | - Erin N Dankert
- The Hormel Institute, University of Minnesota, Austin, MN, 55912
| | - Sk Kayum Alam
- The Hormel Institute, University of Minnesota, Austin, MN, 55912
| | - Luke H Hoeppner
- The Hormel Institute, University of Minnesota, Austin, MN, 55912
| |
Collapse
|
16
|
Innovative Disease Model: Zebrafish as an In Vivo Platform for Intestinal Disorder and Tumors. Biomedicines 2017; 5:biomedicines5040058. [PMID: 28961226 PMCID: PMC5744082 DOI: 10.3390/biomedicines5040058] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Revised: 09/26/2017] [Accepted: 09/29/2017] [Indexed: 12/12/2022] Open
Abstract
Colorectal cancer (CRC) is one of the world’s most common cancers and is the second leading cause of cancer deaths, causing more than 50,000 estimated deaths each year. Several risk factors are highly associated with CRC, including being overweight, eating a diet high in red meat and over-processed meat, having a history of inflammatory bowel disease, and smoking. Previous zebrafish studies have demonstrated that multiple oncogenes and tumor suppressor genes can be regulated through genetic or epigenetic alterations. Zebrafish research has also revealed that the activation of carcinogenesis-associated signal pathways plays an important role in CRC. The biology of cancer, intestinal disorders caused by carcinogens, and the morphological patterns of tumors have been found to be highly similar between zebrafish and humans. Therefore, the zebrafish has become an important animal model for translational medical research. Several zebrafish models have been developed to elucidate the characteristics of gastrointestinal diseases. This review article focuses on zebrafish models that have been used to study human intestinal disorders and tumors, including models involving mutant and transgenic fish. We also report on xenograft models and chemically-induced enterocolitis. This review demonstrates that excellent zebrafish models can provide novel insights into the pathogenesis of gastrointestinal diseases and help facilitate the evaluation of novel anti-tumor drugs.
Collapse
|
17
|
Shaikho S, Dobson CC, Naing T, Samanfar B, Moteshareie H, Hajikarimloo M, Golshani A, Holcik M. Elevated levels of ribosomal proteins eL36 and eL42 control expression of Hsp90 in rhabdomyosarcoma. ACTA ACUST UNITED AC 2016; 4:e1244395. [PMID: 28090422 DOI: 10.1080/21690731.2016.1244395] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Revised: 09/13/2016] [Accepted: 09/28/2016] [Indexed: 01/08/2023]
Abstract
Mammalian 90 kDa heat shock protein (Hsp90) is a ubiquitous molecular chaperone whose expression is selectively upregulated during stress, although the precise control mechanism of this increase is yet to be fully elucidated. We used polysome profiling to show that Hsp90α mRNA is selectively translated, while global translation is inhibited during heat stress. Furthermore, we have identified 2 ribosomal proteins, eL36 and eL42 that modulate Hsp90α expression under both normal and heat shock conditions. Importantly, we noted that expression of eL36 and eL42 is elevated in a panel of human rhabdomyosarcomas where it drives high expression of Hsp90 and modulates sensitivity of these cells to an Hsp90 inhibitor 17-AAG.
Collapse
Affiliation(s)
- Sarah Shaikho
- Molecular Biomedicine Program, Children's Hospital of Eastern Ontario Research Institute , Ottawa, Ontario, Canada
| | - Christine C Dobson
- Molecular Biomedicine Program, Children's Hospital of Eastern Ontario Research Institute , Ottawa, Ontario, Canada
| | - Thet Naing
- Molecular Biomedicine Program, Children's Hospital of Eastern Ontario Research Institute , Ottawa, Ontario, Canada
| | - Bahram Samanfar
- Department of Biology and Ottawa Institute of Systems Biology, Carleton University , Ottawa, Ontario, Canada
| | - Houman Moteshareie
- Department of Biology and Ottawa Institute of Systems Biology, Carleton University , Ottawa, Ontario, Canada
| | - Maryam Hajikarimloo
- Department of Biology and Ottawa Institute of Systems Biology, Carleton University , Ottawa, Ontario, Canada
| | - Ashkan Golshani
- Department of Biology and Ottawa Institute of Systems Biology, Carleton University , Ottawa, Ontario, Canada
| | - Martin Holcik
- Molecular Biomedicine Program, Children's Hospital of Eastern Ontario Research Institute , Ottawa, Ontario, Canada
| |
Collapse
|
18
|
Selenoprotein H is an essential regulator of redox homeostasis that cooperates with p53 in development and tumorigenesis. Proc Natl Acad Sci U S A 2016; 113:E5562-71. [PMID: 27588899 DOI: 10.1073/pnas.1600204113] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Selenium, an essential micronutrient known for its cancer prevention properties, is incorporated into a class of selenocysteine-containing proteins (selenoproteins). Selenoprotein H (SepH) is a recently identified nucleolar oxidoreductase whose function is not well understood. Here we report that seph is an essential gene regulating organ development in zebrafish. Metabolite profiling by targeted LC-MS/MS demonstrated that SepH deficiency impairs redox balance by reducing the levels of ascorbate and methionine, while increasing methionine sulfoxide. Transcriptome analysis revealed that SepH deficiency induces an inflammatory response and activates the p53 pathway. Consequently, loss of seph renders larvae susceptible to oxidative stress and DNA damage. Finally, we demonstrate that seph interacts with p53 deficiency in adulthood to accelerate gastrointestinal tumor development. Overall, our findings establish that seph regulates redox homeostasis and suppresses DNA damage. We hypothesize that SepH deficiency may contribute to the increased cancer risk observed in cohorts with low selenium levels.
Collapse
|
19
|
Ceol CJ, Houvras Y. Uncharted Waters: Zebrafish Cancer Models Navigate a Course for Oncogene Discovery. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 916:3-19. [PMID: 27165347 DOI: 10.1007/978-3-319-30654-4_1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Over a decade has elapsed since the first genetically-engineered zebrafish cancer model was described. During this time remarkable progress has been made. Sophisticated genetic tools have been built to generate oncogene expressing cancers and characterize multiple models of solid and blood tumors. These models have led to unique insights into mechanisms of tumor initiation and progression. New drug targets have been identified, particularly through the functional analysis of cancer genomes. Now in the second decade, zebrafish cancer models are poised for even faster growth as they are used in high-throughput genetic analyses to elucidate key mechanisms underlying critical cancer phenotypes.
Collapse
Affiliation(s)
- Craig J Ceol
- Program in Molecular Medicine and Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, 368 Plantation Street, Worcester, MA, 01605, USA.
| | - Yariv Houvras
- Departments of Surgery and Medicine, Weill Cornell Medical College, 1300 York Avenue, New York, NY, 10065, USA.
| |
Collapse
|
20
|
Mayrhofer M, Mione M. The Toolbox for Conditional Zebrafish Cancer Models. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 916:21-59. [PMID: 27165348 DOI: 10.1007/978-3-319-30654-4_2] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Here we describe the conditional zebrafish cancer toolbox, which allows for fine control of the expression of oncogenes or downregulation of tumor suppressors at the spatial and temporal level. Methods such as the Gal4/UAS or the Cre/lox systems paved the way to the development of elegant tumor models, which are now being used to study cancer cell biology, clonal evolution, identification of cancer stem cells and anti-cancer drug screening. Combination of these tools, as well as novel developments such as the promising genome editing system through CRISPR/Cas9 and clever application of light reactive proteins will enable the development of even more sophisticated zebrafish cancer models. Here, we introduce this growing toolbox of conditional transgenic approaches, discuss its current application in zebrafish cancer models and provide an outlook on future perspectives.
Collapse
Affiliation(s)
- Marie Mayrhofer
- Institute of Toxicology and Genetics, Karlsruhe Institute of Technology, Eggenstein-Leopoldshafen, Germany
| | - Marina Mione
- Institute of Toxicology and Genetics, Karlsruhe Institute of Technology, Eggenstein-Leopoldshafen, Germany.
| |
Collapse
|
21
|
den Hertog J. Tumor Suppressors in Zebrafish: From TP53 to PTEN and Beyond. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 916:87-101. [PMID: 27165350 DOI: 10.1007/978-3-319-30654-4_4] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Zebrafish are increasingly being used to study cancer. Almost all tumor types have been found in zebrafish. However, tumor incidence is relatively low and tumors develop late in life. Functional inactivation of tumor suppressors is a crucial step in cancer progression and more and more tumor suppressor genes are being studied in zebrafish. Most often tumor suppressors have been inactivated by reverse genetics approaches using targeted disruption. However, some tumor suppressor mutants were identified by forward genetic screens for mutants with a particular phenotype. Some of the latter genes had not been recognized as tumor suppressors yet. Similarly, a screen for genes that suppress tumor formation in zebrafish in vivo led to the identification of a novel tumor suppressor gene. In this review, I will provide an overview of what the zebrafish has taught us about tumor suppressors.
Collapse
Affiliation(s)
- Jeroen den Hertog
- Hubrecht Institute-KNAW and University Medical Center Utrecht, Uppsalalaan 8, 3584 CT, Utrecht, The Netherlands. .,Institute of Biology, Leiden University, 2300 RA, Leiden, The Netherlands.
| |
Collapse
|
22
|
Nassar D, Latil M, Boeckx B, Lambrechts D, Blanpain C. Genomic landscape of carcinogen-induced and genetically induced mouse skin squamous cell carcinoma. Nat Med 2015; 21:946-54. [PMID: 26168291 DOI: 10.1038/nm.3878] [Citation(s) in RCA: 146] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2014] [Accepted: 05/18/2015] [Indexed: 12/17/2022]
Abstract
Mouse models of cancers are routinely used to study cancer biology. However, it remains unclear whether carcinogenesis in mice is driven by the same spectrum of genomic alterations found in humans. Here we conducted a comprehensive genomic analysis of 9,10-dimethyl-1,2-benzanthracene (DMBA)-induced skin cancer, the most commonly used skin cancer model, which appears as benign papillomas that progress into squamous cell carcinomas (SCCs). We also studied genetically induced SCCs that expressed G12D mutant Kras (Kras G12D) but were deficient for p53. Using whole-exome sequencing, we uncovered a characteristic mutational signature of DMBA-induced SCCs. We found that the vast majority of DMBA-induced SCCs presented recurrent mutations in Hras, Kras or Rras2 and mutations in several additional putative oncogenes and tumor-suppressor genes. Similar genes were recurrently mutated in mouse and human SCCs that were from different organs or had been exposed to different carcinogens. Invasive SCCs, but not papillomas, presented substantial chromosomal aberrations, especially in DMBA-induced and genetically induced Trp53-mutated SCCs. Metastasis occurred through sequential spreading, with relatively few additional genetic events. This study provides a framework for future functional cancer genomic studies in mice.
Collapse
Affiliation(s)
- Dany Nassar
- Institut de recherche interdisciplinaire en biologie humaine et moléculaire (IRIBHM), Université libre de Buxelles (ULB), Brussels, Belgium
| | - Mathilde Latil
- Institut de recherche interdisciplinaire en biologie humaine et moléculaire (IRIBHM), Université libre de Buxelles (ULB), Brussels, Belgium
| | - Bram Boeckx
- 1] Vesalius Research Center, Vlaams Instituut voor Biotechnologie VIB, Leuven, Belgium. [2] Laboratory for Translational Genetics, Department of Oncology, Katholieke Universiteit Leuven (KUL), Leuven, Belgium
| | - Diether Lambrechts
- 1] Vesalius Research Center, Vlaams Instituut voor Biotechnologie VIB, Leuven, Belgium. [2] Laboratory for Translational Genetics, Department of Oncology, Katholieke Universiteit Leuven (KUL), Leuven, Belgium
| | - Cédric Blanpain
- 1] Institut de recherche interdisciplinaire en biologie humaine et moléculaire (IRIBHM), Université libre de Buxelles (ULB), Brussels, Belgium. [2] WELBIO, Brussels, Belgium
| |
Collapse
|