1
|
Pacheco-Castillo NC, Gómez-Montalvo J, Olivares-Illana V, Recillas-Targa F, Tokar EJ, Avendaño-Vázquez SE, Escudero-Lourdes C. Inorganic Arsenic Induces Elevated p53 Levels with Altered Functionality Impacting the Expression of Toll-like Receptor 3 and Other Target Genes in Immortalized Prostate Epithelial Cells. Int J Mol Sci 2025; 26:4253. [PMID: 40362489 PMCID: PMC12072582 DOI: 10.3390/ijms26094253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2025] [Revised: 04/18/2025] [Accepted: 04/24/2025] [Indexed: 05/15/2025] Open
Abstract
Prostate cancer (PCa) is a major global health concern, particularly in advanced stages where chemotherapy resistance and androgen-independent tumor growth reduce survival rates to below 30%. Toll-like receptor 3 (TLR3), regulated by tumor suppressor p53, is a promising therapeutic target due to its role in tumor cell apoptosis. However, chronic exposure to inorganic arsenic (iAs), a known carcinogen, has been linked to PCa progression and reduced TLR3 expression and activation by polyinosinic/polycytidylic acid (Poly(I/C)), a synthetic ligand used in PCa immunotherapy. Here, we demonstrate that chronic sodium arsenite (NaAsO) exposure increases p53 transcript and protein levels in immortalized prostate epithelial cells. Despite this, key p53 target genes, including TLR3, CDKN1A, and BAX, were significantly downregulated, indicating a transcriptionally inactive p53. Chromatin immunoprecipitation (ChIP) confirmed diminished p53 binding to TLR3 and CDKN1A promoters, while sequencing ruled out TP53 mutations. A bioinformatic analysis revealed elevated TP53 but reduced TLR3 and CDKN1A in prostate adenocarcinoma, suggesting that iAs-induced oxidative stress disrupts p53 function. These findings reveal a novel mechanism by which iAs promotes PCa progression through impaired p53 activity, highlighting the need to explore post-translational and epigenetic factors affecting p53. Restoring p53 transcriptional activity may offer a therapeutic strategy for PCa patients exposed to NaAsO.
Collapse
Affiliation(s)
- Nancy C. Pacheco-Castillo
- Laboratorio de Inmunotoxicología, Facultad de Ciencias Químicas, Universidad Autónoma de San Luis Potosí, San Luis Potosí 78210, Mexico;
| | - Jesús Gómez-Montalvo
- Consorcio de RNA, Laboratorio de Metabolismo de RNA Largos y Medicina Molecular, Instituto Nacional de Medicina Genómica, Ciudad de México 14610, Mexico;
| | - Vanesa Olivares-Illana
- Laboratorio de Interacciones Biomoleculares y Cáncer, Instituto de Física, Universidad Autónoma de San Luis Potosí, San Luis Potosí 78210, Mexico;
| | - Félix Recillas-Targa
- Instituto de Fisiología Celular, Departamento de Genética Molecular, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico;
| | - Erik J. Tokar
- Stem Cell Toxicology Group, Division of Translational Toxicology, National Institute of Environmental Health Science, Durham, NC 27709, USA;
| | - S. Eréndira Avendaño-Vázquez
- Consorcio de RNA, Laboratorio de Metabolismo de RNA Largos y Medicina Molecular, Instituto Nacional de Medicina Genómica, Ciudad de México 14610, Mexico;
| | - Claudia Escudero-Lourdes
- Laboratorio de Inmunotoxicología, Facultad de Ciencias Químicas, Universidad Autónoma de San Luis Potosí, San Luis Potosí 78210, Mexico;
| |
Collapse
|
2
|
Zhang HY, Zhou XQ, Jiang WD, Wu P, Liu Y, Ren HM, Jin XW, Zhang RN, Li H, Mi HF, Zhang L, Feng L. Reversing Zearalenone Toxicity: The Role of Hydroxytyrosol in Zebrafish. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:25905-25918. [PMID: 39530315 DOI: 10.1021/acs.jafc.4c05615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
Zearalenone (ZEA) is a widely distributed mycotoxin that presents a substantial worldwide health risk to animals. Several natural compounds have shown promise in mitigating the detrimental impacts of ZEA. This study examined the detoxification potential of previously identified compounds by utilizing zebrafish embryos as a model organism. Hydroxytyrosol stands out among these natural compounds. Our findings indicate that hydroxytyrosol effectively mitigated mortality, hatching delay, and phenotypic abnormalities induced by ZEA in the assessed embryos. Furthermore, hydroxytyrosol restored the frequency and intensity of tail coiling (TC) while decreasing the expression of heat shock proteins (HSPs) in the zebrafish embryos. Extended incubation with hydroxytyrosol demonstrated protective effects on zebrafish growth and morphology, muscle birefringence, and touch-evoked escape behavior. Subsequent investigations indicated that hydroxytyrosol reversed the expression of proapoptotic targets (e.g., bax and caspase8) and cell cycle regulators (e.g., p21, gadd45a, and rbl2), thereby mitigating apoptosis and G2 cell cycle arrest induced by ZEA in zebrafish embryos. Additionally, hydroxytyrosol decreased staining for senescence associated-β-galactosidase (SA-β-Gal). Notably, p53/FoxO pathway plays an important role in detoxification mechanisms. Overall, these novel findings highlight the potential of hydroxytyrosol to reverse ZEA-induced toxicity in multiple aspects. The mitigating effect of hydroxytyrosol on ZEA toxicity may have been underestimated.
Collapse
Affiliation(s)
- Hong-Yun Zhang
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China
| | - Xiao-Qiu Zhou
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China
- Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
- Key Laboratory for Animal Disease-Resistance Nutrition, Chengdu 611130, China
| | - Wei-Dan Jiang
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China
- Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
- Key Laboratory of Animal Disease-Resistant Nutrition, Ministry of Education, Chengdu 611130, China
| | - Pei Wu
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China
- Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
- Key Laboratory of Animal Disease-Resistant Nutrition, Ministry of Education, Chengdu 611130, China
| | - Yang Liu
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China
- Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
- Key Laboratory of Animal Disease-Resistant Nutrition, Ministry of Education, Chengdu 611130, China
- Key Laboratory of Animal Disease-Resistant Nutrition and Feed, Ministry of Agriculture and Rural Affairs, Chengdu 611130, China
| | - Hong-Mei Ren
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China
| | - Xiao-Wan Jin
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China
| | - Rui-Nan Zhang
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China
| | - Hua Li
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China
| | - Hai-Feng Mi
- Tongwei Research Institute, Chengdu 600438, China
| | - Lu Zhang
- Tongwei Research Institute, Chengdu 600438, China
| | - Lin Feng
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China
- Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
- Key Laboratory for Animal Disease-Resistance Nutrition, Chengdu 611130, China
| |
Collapse
|
3
|
Ren XG, Li W, Li WX, Yu WQ. Mechanism of Histone Arginine Methylation Dynamic Change in Cellular Stress. Int J Mol Sci 2024; 25:7562. [PMID: 39062806 PMCID: PMC11277302 DOI: 10.3390/ijms25147562] [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: 05/08/2024] [Revised: 06/03/2024] [Accepted: 06/29/2024] [Indexed: 07/28/2024] Open
Abstract
Histone arginine residue methylation is crucial for individual development and gene regulation. However, the dynamics of histone arginine methylation in response to cellular stress remains largely unexplored. In addition, the interplay and regulatory mechanisms between this and other histone modifications are important scientific questions that require further investigation. This study aimed to investigate the changes in histone arginine methylation in response to DNA damage. We report a global decrease in histone H3R26 symmetric dimethylation (H3R26me2s) and hypoacetylation at the H3K27 site in response to DNA damage. Notably, H3R26me2s exhibits a distribution pattern similar to that of H3K27ac across the genome, both of which are antagonistic to H3K27me3. Additionally, histone deacetylase 1 (HDAC1) may be recruited to the H3R26me2s demethylation region to mediate H3K27 deacetylation. These findings suggest crosstalk between H3R26me2s and H3K27ac in regulating gene expression.
Collapse
Affiliation(s)
| | | | | | - Wen-Qiang Yu
- Department of RNA Epigenetics, Faculty of Institute of Biomedical Sciences, Campus of Shanghai Medical College, Fudan University, Shanghai 200032, China; (X.-G.R.); (W.L.); (W.-X.L.)
| |
Collapse
|
4
|
Carvalho MFL, Calicchio CS, de Almeida BO, de Miranda LBL, Lipreri da Silva JC, Lima K, Machado-Neto JA. Transcriptomics analysis identified ezrin as a potential druggable target in cervical and gastric cancer cells. Clinics (Sao Paulo) 2024; 79:100422. [PMID: 38972247 PMCID: PMC11276928 DOI: 10.1016/j.clinsp.2024.100422] [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: 02/01/2024] [Revised: 05/13/2024] [Accepted: 06/10/2024] [Indexed: 07/09/2024] Open
Abstract
OBJECTIVE Cancer genomics and transcriptomics studies have provided a large volume of data that enables to test of hypotheses based on real data from cancer patients. Ezrin (encoded by the EZR gene) is a highly expressed protein in cancer that contributes to linking the actin cytoskeleton to the cell membrane and signal transduction pathways involved in oncogenesis and disease progression. NSC305787 is a pharmacological ezrin inhibitor with potential antineoplastic effects. In the present study, the authors prospected EZR mRNA levels in a pan-cancer analysis and identified potential cancers that could benefit from anti-EZR therapies. METHODS This study analyzed TCGA data for 32 cancer types, emphasizing cervical squamous cell carcinoma and stomach adenocarcinoma. It investigated the impact of EZR transcript levels on clinical outcomes and identified differentially expressed genes. Cell lines were treated with NSC305787, and its effects were assessed through various cellular and molecular assays. RESULTS EZR mRNA levels are highly expressed, and their expression is associated with biologically relevant molecular processes in cervical squamous carcinoma and stomach adenocarcinoma. In cellular models of cervical and gastric cancer, NSC305787 reduces cell viability and clonal growth (p < 0.05). Molecular analyses indicate that the pharmacological inhibition of EZR induces molecular markers of cell death and DNA damage, in addition, to promoting the expression of genes associated with apoptosis and inhibiting the expression of genes related to survival and proliferation. CONCLUSION The present findings provide promising evidence that ezrin may be a molecular target in the treatment of cervical and gastric carcinoma.
Collapse
Affiliation(s)
| | - Carolina Santana Calicchio
- Department of Pharmacology, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Bruna Oliveira de Almeida
- Department of Pharmacology, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, SP, Brazil
| | | | | | - Keli Lima
- Department of Pharmacology, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, SP, Brazil; Laboratory of Medical Investigation in Pathogenesis and Targeted Therapy in Onco-Immuno-Hematology (LIM-31), Department of Internal Medicine, Hematology Division, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brazil
| | - João Agostinho Machado-Neto
- Department of Pharmacology, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, SP, Brazil.
| |
Collapse
|
5
|
Du D, Zhou M, Ju C, Yin J, Wang C, Xu X, Yang Y, Li Y, Cui L, Wang Z, Lei Y, Li H, He F, He J. METTL1-mediated tRNA m 7G methylation and translational dysfunction restricts breast cancer tumorigenesis by fueling cell cycle blockade. J Exp Clin Cancer Res 2024; 43:154. [PMID: 38822363 PMCID: PMC11140866 DOI: 10.1186/s13046-024-03076-x] [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: 03/18/2024] [Accepted: 05/20/2024] [Indexed: 06/03/2024] Open
Abstract
BACKGROUND RNA modifications of transfer RNAs (tRNAs) are critical for tRNA function. Growing evidence has revealed that tRNA modifications are related to various disease processes, including malignant tumors. However, the biological functions of methyltransferase-like 1 (METTL1)-regulated m7G tRNA modifications in breast cancer (BC) remain largely obscure. METHODS The biological role of METTL1 in BC progression were examined by cellular loss- and gain-of-function tests and xenograft models both in vitro and in vivo. To investigate the change of m7G tRNA modification and mRNA translation efficiency in BC, m7G-methylated tRNA immunoprecipitation sequencing (m7G tRNA MeRIP-seq), Ribosome profiling sequencing (Ribo-seq), and polysome-associated mRNA sequencing were performed. Rescue assays were conducted to decipher the underlying molecular mechanisms. RESULTS The tRNA m7G methyltransferase complex components METTL1 and WD repeat domain 4 (WDR4) were down-regulated in BC tissues at both the mRNA and protein levels. Functionally, METTL1 inhibited BC cell proliferation, and cell cycle progression, relying on its enzymatic activity. Mechanistically, METTL1 increased m7G levels of 19 tRNAs to modulate the translation of growth arrest and DNA damage 45 alpha (GADD45A) and retinoblastoma protein 1 (RB1) in a codon-dependent manner associated with m7G. Furthermore, in vivo experiments showed that overexpression of METTL1 enhanced the anti-tumor effectiveness of abemaciclib, a cyclin-dependent kinases 4 and 6 (CDK4/6) inhibitor. CONCLUSION Our study uncovered the crucial tumor-suppressive role of METTL1-mediated tRNA m7G modification in BC by promoting the translation of GADD45A and RB1 mRNAs, selectively blocking the G2/M phase of the cell cycle. These findings also provided a promising strategy for improving the therapeutic benefits of CDK4/6 inhibitors in the treatment of BC patients.
Collapse
Affiliation(s)
- Dan Du
- Department of Medical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Mingxia Zhou
- Department of Gastroenterology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Chenxi Ju
- Department of Medical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Jie Yin
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Chang Wang
- Department of Medical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Xinyu Xu
- Department of Medical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Yunqing Yang
- Department of Breast Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Yun Li
- Department of Breast Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Le Cui
- Department of Breast Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Zhengyang Wang
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Yuqing Lei
- Department of Molecular Pathology, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, 450008, China
| | - Hongle Li
- Department of Molecular Pathology, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, 450008, China.
| | - Fucheng He
- Department of Medical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
| | - Jing He
- Department of Breast Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
| |
Collapse
|
6
|
Drake DM, Afsharian K, Or B, Shapiro AM, Lai ML, Miller L, Wells PG. BRCA1 protein dose-dependent risk for embryonic oxidative DNA damage, embryopathies and neurodevelopmental disorders with and without ethanol exposure. Redox Biol 2024; 70:103070. [PMID: 38359745 PMCID: PMC10877410 DOI: 10.1016/j.redox.2024.103070] [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: 01/16/2024] [Accepted: 01/30/2024] [Indexed: 02/17/2024] Open
Abstract
Although widely known as a tumor suppressor, the breast cancer 1 susceptibility protein (BRCA1) is also important in development, where it regulates fetal DNA repair pathways that protect against DNA damage caused by physiological and drug-enhanced levels of reactive oxygen species (ROS). We previously showed that conditional heterozygous (+/-) knockout (cKO) mouse embryos with a minor 28% BRCA1 deficiency developed normally in culture, but when exposed to the ROS-initiating drug, alcohol (ethanol, EtOH), exhibited embryopathies not evident in wild-type (+/+) littermates. Herein, we characterized a directBrca1 +/- knockout (KO) model with a 2-fold greater (58%) reduction in BRCA1 protein vs. the cKO model. We also characterized and compared learning & memory deficits in both the cKO and KO models. Even saline-exposed Brca1 +/- vs. +/+ KO progeny exhibited enhanced oxidative DNA damage and embryopathies in embryo culture and learning & memory deficits in females in vivo, which were not observed in the cKO model, revealing the potential pathogenicity of physiological ROS levels. The embryopathic EtOH concentration for cultured direct KO embryos was half that for cKO embryos, and EtOH affected Brca1 +/+ embryos only in the direct KO model. The spectrum and severity of EtOH embryopathies in culture were greater in both Brca1 +/- vs. +/+ embryos, and direct KO vs. cKO +/- embryos. Motor coordination deficits were evident in both male and female Brca1 +/- KO progeny exposed in utero to EtOH. The results in our direct KO model with a greater BRCA1 deficiency vs. cKO mice provide the first evidence for BRCA1 protein dose-dependent susceptibility to developmental disorders caused by physiological and drug-enhanced oxidative stress.
Collapse
Affiliation(s)
- Danielle M Drake
- Department of Pharmaceutical Sciences and Centre for Pharmaceutical Oncology, Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
| | - Kian Afsharian
- Department of Pharmaceutical Sciences and Centre for Pharmaceutical Oncology, Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
| | - Benjamin Or
- Department of Pharmacology and Toxicology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Aaron M Shapiro
- Department of Pharmaceutical Sciences and Centre for Pharmaceutical Oncology, Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
| | - Michelle L Lai
- Department of Pharmacology and Toxicology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Lutfiya Miller
- Department of Pharmacology and Toxicology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Peter G Wells
- Department of Pharmaceutical Sciences and Centre for Pharmaceutical Oncology, Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada; Department of Pharmacology and Toxicology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
7
|
Shannon MJ, McNeill GL, Koksal B, Baltayeva J, Wächter J, Castellana B, Peñaherrera MS, Robinson WP, Leung PCK, Beristain AG. Single-cell assessment of primary and stem cell-derived human trophoblast organoids as placenta-modeling platforms. Dev Cell 2024; 59:776-792.e11. [PMID: 38359834 DOI: 10.1016/j.devcel.2024.01.023] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 11/15/2023] [Accepted: 01/23/2024] [Indexed: 02/17/2024]
Abstract
Human trophoblast stem cells (hTSCs) and related trophoblast organoids are state-of-the-art culture systems that facilitate the study of trophoblast development and human placentation. Using single-cell transcriptomics, we evaluate how organoids derived from freshly isolated first-trimester trophoblasts or from established hTSC cell lines reproduce developmental cell trajectories and transcriptional regulatory processes defined in vivo. Although organoids from primary trophoblasts and hTSCs overall model trophoblast differentiation with accuracy, specific features related to trophoblast composition, trophoblast differentiation, and transcriptional drivers of trophoblast development show levels of misalignment. This is best illustrated by the identification of an expanded progenitor state in stem cell-derived organoids that is nearly absent in vivo and transcriptionally shares both villous cytotrophoblast and extravillous trophoblast characteristics. Together, this work provides a comprehensive resource that identifies strengths and limitations of current trophoblast organoid platforms.
Collapse
Affiliation(s)
- Matthew J Shannon
- The British Columbia Children's Hospital Research Institute, Vancouver, BC, Canada; Department of Obstetrics & Gynecology, The University of British Columbia, Vancouver, BC, Canada
| | - Gina L McNeill
- The British Columbia Children's Hospital Research Institute, Vancouver, BC, Canada; Department of Obstetrics & Gynecology, The University of British Columbia, Vancouver, BC, Canada
| | - Burak Koksal
- The British Columbia Children's Hospital Research Institute, Vancouver, BC, Canada; Department of Obstetrics & Gynecology, The University of British Columbia, Vancouver, BC, Canada
| | - Jennet Baltayeva
- The British Columbia Children's Hospital Research Institute, Vancouver, BC, Canada; Department of Obstetrics & Gynecology, The University of British Columbia, Vancouver, BC, Canada
| | - Jasmin Wächter
- Department of Obstetrics and Gynecology, Medical University of Vienna, Vienna, Austria
| | - Barbara Castellana
- The British Columbia Children's Hospital Research Institute, Vancouver, BC, Canada; Department of Obstetrics & Gynecology, The University of British Columbia, Vancouver, BC, Canada
| | - Maria S Peñaherrera
- The British Columbia Children's Hospital Research Institute, Vancouver, BC, Canada; Department of Medical Genetics, The University of British Columbia, Vancouver, BC, Canada
| | - Wendy P Robinson
- The British Columbia Children's Hospital Research Institute, Vancouver, BC, Canada; Department of Medical Genetics, The University of British Columbia, Vancouver, BC, Canada
| | - Peter C K Leung
- The British Columbia Children's Hospital Research Institute, Vancouver, BC, Canada; Department of Obstetrics & Gynecology, The University of British Columbia, Vancouver, BC, Canada
| | - Alexander G Beristain
- The British Columbia Children's Hospital Research Institute, Vancouver, BC, Canada; Department of Obstetrics & Gynecology, The University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
8
|
Chen L, Fang C, Yuan X, Liu M, Wu P, Zhong L, Chen Z. Has-miR-300-GADD45B promotes melanoma growth via cell cycle. Aging (Albany NY) 2023; 15:13920-13943. [PMID: 38070141 PMCID: PMC10756120 DOI: 10.18632/aging.205276] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Accepted: 10/16/2023] [Indexed: 12/21/2023]
Abstract
Response to oncogenic factors like UV, GADD45 family in skin participates in scavenging ROS, DNA repair and cell cycle control. Because of this, the previous study of the chronic UVB injury model has found that hsa-miR-300 can conduct intercellular transport by exosomes and target regulation of GADD45B. Whether the hsa-miR-300-GADD45B still regulates tumor development by cell cycle pathway is unclear. Through transcriptomic analysis of primary (n=39) and metastatic (n=102) melanoma, it was confirmed that in metastatic samples, some of the 97 down-regulated genes participate in maintaining skin homeostasis while 42 up-regulated genes were enriched in cancer-related functions. Furthermore, CDKN1A, CDKN2A, CXCR4 and RAD51 in the melanoma pathway, were also differentially expressed between normal skin and melanoma. CDKN1A and CDKN2A were also found to be involved in TP53-dependent cell cycle regulation. In conclusion, it was speculated that CDKN1A, CDKN2A, TP53, GADD45B and hsa-miR-300 may have regulatory relationships. It was demonstrated that there is a bidirectional regulation between hsa-miR-300 and TP53. In addition, miR-300 can regulate CDKN1A by GADD45B/TP53 and promote melanoma growth by accelerating the cell cycle transition from G1/S to G2 phase.
Collapse
Affiliation(s)
- Long Chen
- Department of Burn Plastic and Cosmetology, Affiliated Fuling Hospital, Chongqing University, Chongqing 408099, China
- College of Bioengineering, Chongqing University, Chongqing 400000, China
- Department of Immunology, School of Basic Medical Sciences, Chengdu Medical College, Chengdu 610500, Sichuan, China
- Non-Coding RNA and Drug Discovery Key Laboratory of Sichuan Province, Chengdu Medical College, Chengdu 610500, Sichuan, China
| | - Chenglong Fang
- Department of Rehabilitation, LinYi People’s Hospital, Linyi 276000, Shandong, China
| | - Xiaoxue Yuan
- College of Bioengineering, Chongqing University, Chongqing 400000, China
| | - Mengqi Liu
- College of Bioengineering, Chongqing University, Chongqing 400000, China
| | - Ping Wu
- Department of Burn Plastic and Cosmetology, Affiliated Fuling Hospital, Chongqing University, Chongqing 408099, China
| | - Li Zhong
- College of Bioengineering, Chongqing University, Chongqing 400000, China
| | - Zhiyong Chen
- Department of Burn Plastic and Cosmetology, Affiliated Fuling Hospital, Chongqing University, Chongqing 408099, China
- College of Bioengineering, Chongqing University, Chongqing 400000, China
| |
Collapse
|
9
|
Drake DM, Zhen D, Kerrebijn I, Or B, Gao S, Afsharian K, Tran J, Bhatia S, Cheng A, Wells PG. Breast cancer 1 (BRCA1) protection in altered gene expression and neurodevelopmental disorders due to physiological and ethanol-enhanced reactive oxygen species formation. Free Radic Biol Med 2023; 208:272-284. [PMID: 37541454 DOI: 10.1016/j.freeradbiomed.2023.08.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 06/08/2023] [Accepted: 08/02/2023] [Indexed: 08/06/2023]
Abstract
The breast cancer 1 (Brca1) susceptibility gene regulates the repair of reactive oxygen species (ROS)-mediated DNA damage, which is implicated in neurodevelopmental disorders. Alcohol (ethanol, EtOH) exposure during pregnancy causes fetal alcohol spectrum disorders (FASD), including abnormal brain function, associated with enhanced ROS-initiated DNA damage. Herein, oxidative DNA damage in fetal brains and neurodevelopmental disorders were enhanced in saline-exposed +/- vs. +/+ Brca1 littermates. A single EtOH exposure during gestation further enhanced oxidative DNA damage, altered the expression of developmental/DNA damage response genes in fetal brains, and resulted in neurodevelopmental disorders, all of which were BRCA1-dependent. Pretreatment with the ROS inhibitor phenylbutylnitrone (PBN) blocked DNA damage and some neurodevelopmental disorders in both saline- and EtOH-exposed progeny, corroborating a ROS-dependent mechanism. Fetal BRCA1 protects against altered gene expression and neurodevelopmental disorders caused by both physiological and EtOH-enhanced levels of ROS formation. BRCA1 deficiencies may enhance the risk for FASD.
Collapse
Affiliation(s)
- Danielle M Drake
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada; Centre for Pharmaceutical Oncology, Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
| | - Danlin Zhen
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
| | - Isabel Kerrebijn
- Department of Pharmacology and Toxicology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Benjamin Or
- Department of Pharmacology and Toxicology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Sophie Gao
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
| | - Kian Afsharian
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada; Centre for Pharmaceutical Oncology, Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
| | - Jason Tran
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
| | - Shama Bhatia
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada; Centre for Pharmaceutical Oncology, Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
| | - Ashley Cheng
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada; Centre for Pharmaceutical Oncology, Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
| | - Peter G Wells
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada; Centre for Pharmaceutical Oncology, Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada; Department of Pharmacology and Toxicology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
10
|
Darwish T, Swaidan NT, Emara MM. Stress Factors as Possible Regulators of Pluripotent Stem Cell Survival and Differentiation. BIOLOGY 2023; 12:1119. [PMID: 37627003 PMCID: PMC10452095 DOI: 10.3390/biology12081119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 08/02/2023] [Accepted: 08/05/2023] [Indexed: 08/27/2023]
Abstract
In recent years, extensive research efforts have been directed toward pluripotent stem cells, primarily due to their remarkable capacity for pluripotency. This unique attribute empowers these cells to undergo self-renewal and differentiate into various cell types originating from the ectoderm, mesoderm, and endoderm germ layers. The delicate balance and precise regulation of self-renewal and differentiation are essential for the survival and functionality of these cells. Notably, exposure to specific environmental stressors can activate numerous transcription factors, initiating a diverse array of stress response pathways. These pathways play pivotal roles in regulating gene expression and protein synthesis, ultimately aiming to preserve cell survival and maintain cellular functions. Reactive oxygen species, heat shock, hypoxia, osmotic stress, DNA damage, endoplasmic reticulum stress, and mechanical stress are among the examples of such stressors. In this review, we comprehensively discuss the impact of environmental stressors on the growth of embryonic cells. Furthermore, we provide a summary of the distinct stress response pathways triggered when pluripotent stem cells are exposed to different environmental stressors. Additionally, we highlight recent discoveries regarding the role of such stressors in the generation, differentiation, and self-renewal of induced pluripotent stem cells.
Collapse
Affiliation(s)
| | | | - Mohamed M. Emara
- Basic Medical Sciences Department, College of Medicine, QU Health, Qatar University, 2713 Doha, Qatar
| |
Collapse
|
11
|
Begolli R, Chatziangelou M, Samiotaki M, Goutas A, Barda S, Goutzourelas N, Kevrekidis DP, Malea P, Trachana V, Liu M, Lin X, Kollatos N, Stagos D, Giakountis A. Transcriptome and proteome analysis reveals the anti-cancer properties of Hypnea musciformis marine macroalga extract in liver and intestinal cancer cells. Hum Genomics 2023; 17:71. [PMID: 37525271 PMCID: PMC10388463 DOI: 10.1186/s40246-023-00517-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 07/18/2023] [Indexed: 08/02/2023] Open
Abstract
BACKGROUND Marine seaweeds are considered as a rich source of health-promoting compounds by the food and pharmaceutical industry. Hypnea musciformis is a marine red macroalga (seaweed) that is widely distributed throughout the world, including the Mediterranean Sea. It is known to contain various bioactive compounds, including sulfated polysaccharides, flavonoids, and phlorotannins. Recent studies have investigated the potential anticancer effects of extracts from H. musciformis demonstrating their cytotoxic effects on various cancer cell lines. The anticancer effects of these extracts are thought to be due to the presence of bioactive compounds, particularly sulfated polysaccharides, which have been shown to have anticancer and immunomodulatory effects. However, further studies are needed to fully understand the molecular mechanisms that underlie their anticancer effects and to determine their potential as therapeutic agents for cancer treatment. METHODS H. musciformis was collected from the Aegean Sea (Greece) and used for extract preparation. Transcriptome and proteome analysis was performed in liver and colon cancer human cell lines following treatment with H. musciformis seaweed extracts to characterize its anticancer effect in detail at the molecular level and to link transcriptome and proteome responses to the observed phenotypes in cancer cells. RESULTS We have identified that treatment with the seaweed extract triggers a p53-mediated response at the transcriptional and protein level in liver cancer cells, in contrast to colon cancer cells in which the effects are more associated with metabolic changes. Furthermore, we show that in treated HepG2 liver cancer cells, p53 interacts with the chromatin of several target genes and facilitates their upregulation possibly through the recruitment of the p300 co-activator. CONCLUSIONS Overall, the available evidence suggests that extracts from H. musciformis have the potential to serve as a source of anticancer agents in liver cancer cells mainly through activation of a p53-mediated anti-tumor response that is linked to inhibition of cellular proliferation and induction of cell death.
Collapse
Affiliation(s)
- Rodiola Begolli
- Department of Biochemistry and Biotechnology, School of Health Sciences, University of Thessaly, 41500, Biopolis, Larissa, Greece
| | - Myrto Chatziangelou
- Department of Biochemistry and Biotechnology, School of Health Sciences, University of Thessaly, 41500, Biopolis, Larissa, Greece
| | | | - Andreas Goutas
- Department of Biochemistry and Biotechnology, School of Health Sciences, University of Thessaly, 41500, Biopolis, Larissa, Greece
- Department of Biology, Faculty of Medicine, University of Thessaly, 41500, Biopolis, Larissa, Greece
| | - Sofia Barda
- Department of Biochemistry and Biotechnology, School of Health Sciences, University of Thessaly, 41500, Biopolis, Larissa, Greece
| | - Nikolaos Goutzourelas
- Department of Biochemistry and Biotechnology, School of Health Sciences, University of Thessaly, 41500, Biopolis, Larissa, Greece
| | - Dimitrios Phaedon Kevrekidis
- Laboratory of Forensic Medicine and Toxicology, Department of Medicine, Aristotle University of Thessaloniki, 54124, Thessaloniki, Greece
| | - Paraskevi Malea
- Department of Botany, School of Biology, Aristotle University of Thessaloniki, 54124, Thessaloniki, Greece
| | - Varvara Trachana
- Department of Biology, Faculty of Medicine, University of Thessaly, 41500, Biopolis, Larissa, Greece
| | - Ming Liu
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China
- Laboratory for Marine Drugs and Bioproducts of Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266237, China
| | - Xiukun Lin
- College of Marine Sciences, Beibu Gulf University, 12 Binhai Rd, Qinzhou, 535011, Guangxi, China
| | - Nikolaos Kollatos
- Department of Biochemistry and Biotechnology, School of Health Sciences, University of Thessaly, 41500, Biopolis, Larissa, Greece
| | - Dimitrios Stagos
- Department of Biochemistry and Biotechnology, School of Health Sciences, University of Thessaly, 41500, Biopolis, Larissa, Greece.
| | - Antonis Giakountis
- Department of Biochemistry and Biotechnology, School of Health Sciences, University of Thessaly, 41500, Biopolis, Larissa, Greece.
| |
Collapse
|
12
|
Rzepkowska M, Głowacka DK, Szczepkowski M, Fajkowska M. Hepatotoxic effect of dietary phytoestrogens on juvenile cultured Russian sturgeon (Acipenser gueldenstaedtii). AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2023; 261:106639. [PMID: 37515925 DOI: 10.1016/j.aquatox.2023.106639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Revised: 06/23/2023] [Accepted: 07/23/2023] [Indexed: 07/31/2023]
Abstract
In the last two decades, much controversy has grown over the use of soybean products in aquafeeds, especially for carnivorous fish like sturgeons. One point of discussion is the effect of soybean phytoestrogens on fish health. There are many aspects of phytoestrogen utilization in aquafeeds, therefore, the aim of this study is to verify if common legume phytoestrogens can affect juvenile cultured sturgeon erythrocyte and hepatocyte genotoxicity and cause liver pathology. Russian sturgeons were fed from 100 till 365 dph1 with daidzein, genistein, and coumestrol supplemented diets in concentrations: 10, 0.05 and 0.001 g kg-1 of feed, respectively. The SCGE2 method combined with qPCR of three genes involved in DNA repair and genome maintenance, namely cyp1a1, gaad45a and p53 were analyzed. The results were compared with histopathological evaluation of liver tissue. In fish fed with coumestrol supplemented diet, DNA strand damage was the highest in both erythrocytes and hepatocytes, however, simultaneously the lowest level of oxidative DNA damage was found. Additionally, slightly elevated expression of the p53 gene was observed along with a decreased number of apoptotic hepatocytes, which suggests that low concentration of coumestrol may support DNA repair mechanisms in the liver. Although, daidzein showed a preventive effect only against fibrosis. Isoflavones did not show a significant effect on DNA damage in studied cells. Genistein was found to increase macro- and microvesicular steatosis, portal hepatitis and fibrosis, indicating its negative role in the development of liver injuries. Daidzein alleviated some sturgeon liver damage, especially macrovesicular steatosis and interface hepatitis. However, it increased hepatocyte apoptosis, which may suggest daidzein potentially inducing liver injury, though not manifested by other histopathological lesions. Therefore, it can be concluded that at given concentrations, the tested phytoestrogens did not show clearly hepatoprotective effect in sturgeons.
Collapse
Affiliation(s)
- Małgorzata Rzepkowska
- Department of Ichthyology and Biotechnology in Aquaculture, Institute of Animal Sciences, Warsaw University of Life Sciences, 8 Ciszewskiego Street, Warsaw 02-786, Poland.
| | - Dominika Kinga Głowacka
- Department of Ichthyology and Biotechnology in Aquaculture, Institute of Animal Sciences, Warsaw University of Life Sciences, 8 Ciszewskiego Street, Warsaw 02-786, Poland
| | - Mirosław Szczepkowski
- Department of Sturgeon Fish Breeding, National Inland Fisheries Research Institute in Olsztyn, Pieczarki 50, Pozezdrze 11-610, Poland
| | - Magdalena Fajkowska
- Department of Ichthyology and Biotechnology in Aquaculture, Institute of Animal Sciences, Warsaw University of Life Sciences, 8 Ciszewskiego Street, Warsaw 02-786, Poland
| |
Collapse
|
13
|
Zhang Y, Lv M, Jiang H, Li H, Li R, Yang C, Huang Y, Zhou H, Mei Y, Gao J, Cao X. Mitotic defects lead to unreduced sperm formation in cdk1 -/- mutants. Int J Biol Macromol 2023:125171. [PMID: 37271265 DOI: 10.1016/j.ijbiomac.2023.125171] [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: 01/06/2023] [Revised: 05/03/2023] [Accepted: 05/23/2023] [Indexed: 06/06/2023]
Abstract
Unreduced gametes, that are important for species evolution and agricultural development, are generally believed to be formed by meiotic defects. However, we found that male diploid loach (Misgurnus anguillicaudatus) could produce not only haploid sperms, but also unreduced sperms, after cyclin-dependent kinase 1 gene (cdk1, one of the most important kinases in regulating cell mitosis) deletion. Observations on synaptonemal complexes of spermatocyte in prophase of meiosis and spermatogonia suggested that the number of chromosomes in some spermatogonia of cdk1-/- loach doubled, leading to unreduced diploid sperm production. Then, transcriptome analysis revealed aberrant expressions of some cell cycle-related genes (such as ppp1c and gadd45) in spermatogonia of cdk1-/- loach relative to wild-type loach. An in vitro and in vivo experiment further validated that Cdk1 deletion in diploid loach resulted in mitotic defects, leading to unreduced diploid sperm formation. In addition, we found that cdk1-/- zebrafish could also produce unreduced diploid sperms. This study provides important information on revealing the molecular mechanisms behind unreduced gamete formation through mitotic defects, and lays the foundation for a novel strategy for fish polyploidy creation by using cdk1 mutants to produce unreduced sperms, which can then be used to obtain polyploidy, proposed to benefit aquaculture.
Collapse
Affiliation(s)
- Yunbang Zhang
- College of Fisheries, Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Huazhong Agricultural University, Wuhan 430070, China; Hubei Provincial Engineering Laboratory for Pond Aquaculture, Hubei, China
| | - Meiqi Lv
- College of Fisheries, Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Huazhong Agricultural University, Wuhan 430070, China
| | - Hanjun Jiang
- College of Fisheries, Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Huazhong Agricultural University, Wuhan 430070, China
| | - Hui Li
- College of Fisheries, Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Huazhong Agricultural University, Wuhan 430070, China
| | - Rongyun Li
- College of Fisheries, Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Huazhong Agricultural University, Wuhan 430070, China
| | - Chuang Yang
- College of Fisheries, Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Huazhong Agricultural University, Wuhan 430070, China
| | - Yuwei Huang
- College of Fisheries, Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Huazhong Agricultural University, Wuhan 430070, China
| | - He Zhou
- College of Fisheries and Life Science, Dalian Ocean University, Dalian 116023, China
| | - Yihui Mei
- College of Fisheries, Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Huazhong Agricultural University, Wuhan 430070, China
| | - Jian Gao
- College of Fisheries, Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Huazhong Agricultural University, Wuhan 430070, China; Hubei Provincial Engineering Laboratory for Pond Aquaculture, Hubei, China.
| | - Xiaojuan Cao
- College of Fisheries, Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Huazhong Agricultural University, Wuhan 430070, China; Hubei Provincial Engineering Laboratory for Pond Aquaculture, Hubei, China.
| |
Collapse
|
14
|
Kim G, Lee D. Reverse tracking from drug-induced transcriptomes through multilayer molecular networks reveals hidden drug targets. Comput Biol Med 2023; 158:106881. [PMID: 37028141 DOI: 10.1016/j.compbiomed.2023.106881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 03/03/2023] [Accepted: 03/30/2023] [Indexed: 04/03/2023]
Abstract
Identifying molecular targets of a drug is an essential process for drug discovery and development. The recent in-silico approaches are usually based on the structure information of chemicals and proteins. However, 3D structure information is hard to obtain and machine-learning methods using 2D structure suffer from data imbalance problem. Here, we present a reverse tracking method from genes to target proteins using drug-perturbed gene transcriptional profiles and multilayer molecular networks. We scored how well the protein explains gene expression changes perturbed by a drug. We validated the protein scores of our method in predicting known targets of drugs. Our method performs better than other methods using the gene transcriptional profiles and shows the ability to suggest the molecular mechanism of drugs. Furthermore, our method has the potential to predict targets for objects that do not have rigid structural information, such as coronavirus.
Collapse
|
15
|
Friesen A, Fritsch-Decker S, Mülhopt S, Quarz C, Mahl J, Baumann W, Hauser M, Wexler M, Schlager C, Gutmann B, Krebs T, Goßmann AK, Weis F, Hufnagel M, Stapf D, Hartwig A, Weiss C. Comparing the Toxicological Responses of Pulmonary Air-Liquid Interface Models upon Exposure to Differentially Treated Carbon Fibers. Int J Mol Sci 2023; 24:ijms24031927. [PMID: 36768249 PMCID: PMC9915385 DOI: 10.3390/ijms24031927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 01/12/2023] [Accepted: 01/13/2023] [Indexed: 01/20/2023] Open
Abstract
In recent years, the use of carbon fibers (CFs) in various sectors of industry has been increasing. Despite the similarity of CF degradation products to other toxicologically relevant materials such as asbestos fibers and carbon nanotubes, a detailed toxicological evaluation of this class of material has yet to be performed. In this work, we exposed advanced air-liquid interface cell culture models of the human lung to CF. To simulate different stresses applied to CF throughout their life cycle, they were either mechanically (mCF) or thermo-mechanically pre-treated (tmCF). Different aspects of inhalation toxicity as well as their possible time-dependency were monitored. mCFs were found to induce a moderate inflammatory response, whereas tmCF elicited stronger inflammatory as well as apoptotic effects. Furthermore, thermal treatment changed the surface properties of the CF resulting in a presumed adhesion of the cells to the fiber fragments and subsequent cell loss. Triple-cultures encompassing epithelial, macrophage, and fibroblast cells stood out with an exceptionally high inflammatory response. Only a weak genotoxic effect was detected in the form of DNA strand breaks in mono- and co-cultures, with triple-cultures presenting a possible secondary genotoxicity. This work establishes CF fragments as a potentially harmful material and emphasizes the necessity of further toxicological assessment of existing and upcoming advanced CF-containing materials.
Collapse
Affiliation(s)
- Alexandra Friesen
- Karlsruhe Institute of Technology (KIT), Institute of Applied Biosciences, Department of Food Chemistry and Toxicology, 76131 Karlsruhe, Germany
| | - Susanne Fritsch-Decker
- Karlsruhe Institute of Technology (KIT), Institute of Biological and Chemical Systems, Biological Information Processing, 76344 Eggenstein-Leopoldshafen, Germany
| | - Sonja Mülhopt
- Karlsruhe Institute of Technology (KIT), Institute for Technical Chemistry, 76344 Eggenstein-Leopoldshafen, Germany
| | - Caroline Quarz
- Karlsruhe Institute of Technology (KIT), Institute of Applied Biosciences, Department of Food Chemistry and Toxicology, 76131 Karlsruhe, Germany
| | - Jonathan Mahl
- Karlsruhe Institute of Technology (KIT), Institute for Technical Chemistry, 76344 Eggenstein-Leopoldshafen, Germany
| | - Werner Baumann
- Karlsruhe Institute of Technology (KIT), Institute for Technical Chemistry, 76344 Eggenstein-Leopoldshafen, Germany
| | - Manuela Hauser
- Karlsruhe Institute of Technology (KIT), Institute for Technical Chemistry, 76344 Eggenstein-Leopoldshafen, Germany
| | - Manuela Wexler
- Karlsruhe Institute of Technology (KIT), Institute for Technical Chemistry, 76344 Eggenstein-Leopoldshafen, Germany
| | | | | | - Tobias Krebs
- Vitrocell Systems GmbH, 79183 Waldkirch, Germany
| | | | | | - Matthias Hufnagel
- Karlsruhe Institute of Technology (KIT), Institute of Applied Biosciences, Department of Food Chemistry and Toxicology, 76131 Karlsruhe, Germany
| | - Dieter Stapf
- Karlsruhe Institute of Technology (KIT), Institute for Technical Chemistry, 76344 Eggenstein-Leopoldshafen, Germany
| | - Andrea Hartwig
- Karlsruhe Institute of Technology (KIT), Institute of Applied Biosciences, Department of Food Chemistry and Toxicology, 76131 Karlsruhe, Germany
- Correspondence: (A.H.); (C.W.)
| | - Carsten Weiss
- Karlsruhe Institute of Technology (KIT), Institute of Biological and Chemical Systems, Biological Information Processing, 76344 Eggenstein-Leopoldshafen, Germany
- Correspondence: (A.H.); (C.W.)
| |
Collapse
|
16
|
Villa-Jaimes GS, Moshage H, Avelar-González FJ, González-Ponce HA, Buist-Homan M, Guevara-Lara F, Sánchez-Alemán E, Martínez-Hernández SL, Ventura-Juárez J, Muñoz-Ortega MH, Martínez-Saldaña MC. Molecular and Antioxidant Characterization of Opuntia robusta Fruit Extract and Its Protective Effect against Diclofenac-Induced Acute Liver Injury in an In Vivo Rat Model. Antioxidants (Basel) 2023; 12:antiox12010113. [PMID: 36670975 PMCID: PMC9855095 DOI: 10.3390/antiox12010113] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 12/23/2022] [Accepted: 12/27/2022] [Indexed: 01/04/2023] Open
Abstract
A molecular characterization of the main phytochemicals and antioxidant activity of Opuntia robusta (OR) fruit extract was carried out, as well as an evaluation of its hepatoprotective effect against diclofenac (DF)-induced acute liver injury was evaluated. Phenols, flavonoids and betalains were quantified, and antioxidant characterization was performed by means of the ABTS•+, DPPH and FRAP assays. UPLC-QTOF-MS/MS was used to identify the main biocompounds present in OR fruit extract was carried out via. In the in vivo model, groups of rats were treated prophylactically with the OR fruit extract, betanin and N-acteylcysteine followed by a single dose of DF. Biochemical markers of oxidative stress (MDA and GSH) and relative gene expression of the inducible antioxidant response (Nrf2, Sod2, Hmox1, Nqo1 and Gclc), cell death (Casp3) and DNA repair (Gadd45a) were analyzed. Western blot analysis was performed to measure protein levels of Nrf2 and immunohistochemical analysis was used to assess caspase-3 activity in the experimental groups. In our study, the OR fruit extract showed strong antioxidant and cytoprotective capacity due to the presence of bioactive compounds, such as betalain and phenols. We conclude that OR fruit extract or selected components can be used clinically to support patients with acute liver injury.
Collapse
Affiliation(s)
- Gloria Stephanie Villa-Jaimes
- Department de Morfología, Centro de Ciencias Básicas, Universidad Autónoma de Aguascalientes, Aguascalientes 20100, Mexico
| | - Han Moshage
- Department of Gastroenterology and Hepatology, University Medical Center of Groningen, University of Groningen, 9713 ZP Groningen, The Netherlands
- Department of Laboratory Medicine, University Medical Center Groningen, University of Groningen, 9713 ZP Groningen, The Netherlands
| | | | | | - Manon Buist-Homan
- Department of Gastroenterology and Hepatology, University Medical Center of Groningen, University of Groningen, 9713 ZP Groningen, The Netherlands
- Department of Laboratory Medicine, University Medical Center Groningen, University of Groningen, 9713 ZP Groningen, The Netherlands
| | - Fidel Guevara-Lara
- Departamento de Química, Centro de Ciencias Básicas, Universidad Autónoma de Aguascalientes, Aguascalientes 20100, Mexico
| | - Esperanza Sánchez-Alemán
- Department de Morfología, Centro de Ciencias Básicas, Universidad Autónoma de Aguascalientes, Aguascalientes 20100, Mexico
- Unidad de Medicina Familiar 8, Instituto Mexicano del Seguro Social (IMSS), Aguascalientes 20180, Mexico
| | - Sandra Luz Martínez-Hernández
- Departamento de Microbiología, Centro de Ciencias Básicas, Universidad Autónoma de Aguascalientes, Aguascalientes 20100, Mexico
| | - Javier Ventura-Juárez
- Department de Morfología, Centro de Ciencias Básicas, Universidad Autónoma de Aguascalientes, Aguascalientes 20100, Mexico
| | - Martín Humberto Muñoz-Ortega
- Departamento de Química, Centro de Ciencias Básicas, Universidad Autónoma de Aguascalientes, Aguascalientes 20100, Mexico
| | - Ma. Consolación Martínez-Saldaña
- Department de Morfología, Centro de Ciencias Básicas, Universidad Autónoma de Aguascalientes, Aguascalientes 20100, Mexico
- Correspondence:
| |
Collapse
|
17
|
Wang Z, Wen S, Zhong M, Yang Z, Xiong W, Zhang K, Yang S, Li H, Guo S. Epigenetics: Novel crucial approach for osteogenesis of mesenchymal stem cells. J Tissue Eng 2023; 14:20417314231175364. [PMID: 37342486 PMCID: PMC10278427 DOI: 10.1177/20417314231175364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Accepted: 04/26/2023] [Indexed: 06/23/2023] Open
Abstract
Bone has a robust regenerative potential, but its capacity to repair critical-sized bone defects is limited. In recent years, stem cells have attracted significant interest for their potential in tissue engineering. Applying mesenchymal stem cells (MSCs) for enhancing bone regeneration is a promising therapeutic strategy. However, maintaining optimal cell efficacy or viability of MSCs is limited by several factors. Epigenetic modification can cause changes in gene expression levels without changing its sequence, mainly including nucleic acids methylation, histone modification, and non-coding RNAs. This modification is believed to be one of the determinants of MSCs fate and differentiation. Understanding the epigenetic modification of MSCs can improve the activity and function of stem cells. This review summarizes recent advances in the epigenetic mechanisms of MSCs differentiation into osteoblast lineages. We expound that epigenetic modification of MSCs can be harnessed to treat bone defects and promote bone regeneration, providing potential therapeutic targets for bone-related diseases.
Collapse
Affiliation(s)
- Zhaohua Wang
- Department of Plastic Surgery, The First Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Si Wen
- Department of Nephrology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province, China
| | - Meiqi Zhong
- Department of Plastic Surgery, The First Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Ziming Yang
- Department of Plastic Surgery, The First Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Wei Xiong
- Department of Plastic Surgery, The First Hospital of Shihezi University School of Medicine, Shihezi, China
| | - Kuo Zhang
- College of Humanities and Social Sciences, Dalian Medical University, Dalian, Liaoning Province, China
| | - Shude Yang
- Department of Plastic Surgery, The First Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Huizheng Li
- Department of Otorhinolaryngology & Head and Neck Surgery, Dalian Friendship Hospital of Dalian Medical University, Dalian, Liaoning Province, China
| | - Shu Guo
- Department of Plastic Surgery, The First Hospital of China Medical University, Shenyang, Liaoning Province, China
| |
Collapse
|
18
|
Pham DN, Sokolov EP, Falfushynska H, Sokolova IM. Gone with sunscreens: Responses of blue mussels (Mytilus edulis) to a wide concentration range of a UV filter ensulizole. CHEMOSPHERE 2022; 309:136736. [PMID: 36209850 DOI: 10.1016/j.chemosphere.2022.136736] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 09/27/2022] [Accepted: 10/01/2022] [Indexed: 06/16/2023]
Abstract
Organic UV filters have emerged as a new threat to marine organisms, but ecotoxicological studies have so far focused on only a few substances despite the chemical diversity of these synthetic sunscreen agents. Here we examined the responses of blue mussels Mytilus edulis to ensulizole, a non-lipophilic UV filter commonly found in the Baltic Sea. Mussels were exposed for three weeks to five ensulizole concentrations of 10, 102, 103, 104, and 105 ng/L. Stress on stress response was evaluated by subjecting mussels to air exposure. A battery of biomarkers related to detoxification and antioxidant defense, oxidative stress damage, energy reserves and metabolism, autophagy, apoptosis, inflammation, and DNA damage was measured in the gills and the digestive gland. In general, ensulizole affected the antioxidant response, energy storage, and cell death-related processes in mussel tissues. Mussels exposed to low, environmentally relevant concentrations of ensulizole had a shorter air survival time than the control. Ensulizole often showed the non-monotonic concentration-response curves, suggesting the complex effects of this UV filter at molecular, biochemical, and organismal levels.
Collapse
Affiliation(s)
- Duy Nghia Pham
- Department of Marine Biology, Institute of Biological Sciences, University of Rostock, Rostock, Germany
| | - Eugene P Sokolov
- Leibniz Institute for Baltic Sea Research, Leibniz Science Campus Phosphorus Research, Warnemünde, Rostock, Germany
| | - Halina Falfushynska
- Department of Human Health, Physical Rehabilitation and Vital Activity, Ternopil V. Hnatiuk National Pedagogical University, Ternopil, Ukraine
| | - Inna M Sokolova
- Department of Marine Biology, Institute of Biological Sciences, University of Rostock, Rostock, Germany; Department of Maritime Systems, Interdisciplinary Faculty, University of Rostock, Rostock, Germany.
| |
Collapse
|
19
|
Anticancer and chemosensitization effects of cannabidiol in 2D and 3D cultures of TNBC: involvement of GADD45α, integrin-α5, -β5, -β1, and autophagy. Drug Deliv Transl Res 2022; 12:2762-2777. [PMID: 35217991 PMCID: PMC9811521 DOI: 10.1007/s13346-022-01137-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/11/2022] [Indexed: 01/07/2023]
Abstract
To date, promising therapy for triple negative breast cancer (TNBC) remains a serious concern clinically because of poor prognosis, resistance, and recurrence. Herein, anti-cancer potential of synthetic cannabidiol (CBD; Purisys, GA; GMP grade) was explored either alone or as a chemosensitizer followed by post-treatment with doxorubicin (DOX) in TNBC (i.e., MDA-MB-231 and MDA-MB-468) cells. In comparison to 2D cultures, CBD showed greater IC50 values in 3D (LDP2 hydrogel based) cultures of MDA-MB-231 (6.26-fold higher) and MDA-MB-468 (10.22-fold higher) cells. Next-generation RNA sequencing revealed GADD45A, GADD45G, FASN, LOX, and integrin (i.e., -α5, -β5) genes to be novelly altered by CBD in MDA-MB-231 cells. CIM-16 plate-based migration assay and western blotting disclosed that CBD induces anti-migratory effects in TNBC cells by decreasing fibronectin, vimentin, and integrins-α5, -β5, and -β1. Western blotting, RT-qPCR, and immunocytochemistry revealed that CBD inhibited autophagy (decreased Beclin1, and ATG-5, -7, and -16) of TNBC cells. CBD pre-treatment increased DOX sensitivity in TNBC cells. CBD pre-treatment accompanied by DOX treatment decreased LOX and integrin-α5, and increased caspase 9 protein respectively in MDA-MB-468 cells.
Collapse
|
20
|
Rad54L promotes bladder cancer progression by regulating cell cycle and cell senescence. MEDICAL ONCOLOGY (NORTHWOOD, LONDON, ENGLAND) 2022; 39:185. [PMID: 36071250 DOI: 10.1007/s12032-022-01751-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 05/13/2022] [Indexed: 10/14/2022]
Abstract
Bladder cancer (BCa) is the most prevalent cancer of the urinary system, but its pathogenesis is still poorly understood. Several reports have suggested that gene damage repair is highly correlated with tumor development and drug resistance, in which homologous recombination repair gene Rad54L seems to play an important role, through yet unclear mechanisms. Therefore, this study stratified cancer patients by Rad54L expression in BCa tissue, and high Rad54L expression was associated with a poor prognosis. Mechanistically, we demonstrate that high Rad54L expression promotes abnormal bladder tumor cell proliferation by changing the cell cycle and cell senescence. In addition, this study also suggests that Rad54L may be associated with p53, p21, and pRB in BCa tissue. In summary, this study exposes Rad54L as potential a prognostic biomarker and precision treatment target in BCa.
Collapse
|
21
|
Wooten J, Mavingire N, Araújo CA, Aja J, Wauchope S, Delgoda R, Brantley E. Dibenzyl trisulfide induces caspase-independent death and lysosomal membrane permeabilization of triple-negative breast cancer cells. Fitoterapia 2022; 160:105203. [PMID: 35489582 PMCID: PMC9979099 DOI: 10.1016/j.fitote.2022.105203] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 04/21/2022] [Accepted: 04/23/2022] [Indexed: 01/25/2023]
Abstract
The Petiveria alliacea L. (P. alliacea) plant is traditionally used in folklore medicine throughout tropical regions of the world to treat arthritis, asthma, and cancer. Dibenzyl trisulfide (DTS) is one of the active ingredients within the P. alliacea plant. Triple-negative breast cancer (TNBC) is associated with a poor prognosis, particularly among women of West African ancestry, due in part to limited effective therapy. Though potent anticancer actions of DTS have been reported in a TNBC cell line, the mechanism of DTS-mediated cytotoxicity and cell death remains ill-defined. In the current study, we show that DTS exhibits cytotoxicity in a panel of triple-negative breast cancer (TNBC) cells derived from patients of European and West African ancestry. We found that DTS inhibits proliferation and migration of CRL-2335 cells derived from a patient of West African ancestry. DTS induces the expression of pro-apoptotic genes BAK1, GADD45a, and LTA in CRL2335 cells though it primarily promotes caspase-independent CRL-2335 cell death. DTS also promotes destabilization of the lysosomal membrane resulting in cathepsin B release in CRL-2335 cells. Finally, Kaplan-Meier survival curves reveal that higher expression of BAK1 and LTA in tumors from patients with TNBC is associated with longer relapse-free survival. Collectively, our data suggest that DTS confers promising antitumor efficacy in TNBC, in part, via lysosomal-mediated, caspase-independent cell death to warrant furthering its development as an anticancer agent.
Collapse
Affiliation(s)
- Jonathan Wooten
- Department of Basic Sciences, Loma Linda University Health School of Medicine, Loma Linda, CA, United States of America; Center for Health Disparities and Molecular Medicine, Loma Linda University Health School of Medicine, Loma Linda, CA, United States of America.
| | - Nicole Mavingire
- Department of Basic Sciences, Loma Linda University Health School of Medicine, Loma Linda, CA, United States of America.
| | - Cristina A. Araújo
- Department of Basic Sciences, Loma Linda University Health School of Medicine, Loma Linda, CA, US,Center for Health Disparities and Molecular Medicine, Loma Linda University Health School of Medicine, Loma Linda, CA, US
| | - Joyce Aja
- National Institute of Molecular Biology and Biotechnology, University of the Philippines Diliman, Quezon City, Philippines.
| | - Shaniece Wauchope
- Natural Products Institute, University of the West Indies, Mona, Kingston 7, Jamaica.
| | - Rupika Delgoda
- Natural Products Institute, University of the West Indies, Mona, Kingston 7, Jamaica.
| | - Eileen Brantley
- Department of Basic Sciences, Loma Linda University Health School of Medicine, Loma Linda, CA, United States of America; Center for Health Disparities and Molecular Medicine, Loma Linda University Health School of Medicine, Loma Linda, CA, United States of America.
| |
Collapse
|
22
|
Ji Y, He Y, Yang Y, Dai Z, Wu Z. Hydroxyproline alleviates 4-hydroxy-2-nonenal-induced DNA damage and apoptosis in porcine intestinal epithelial cells. ANIMAL NUTRITION 2022; 9:7-15. [PMID: 35949986 PMCID: PMC9344311 DOI: 10.1016/j.aninu.2021.08.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 07/14/2021] [Accepted: 08/08/2021] [Indexed: 11/28/2022]
Abstract
Oxidative stress has been confirmed in relation to intestinal mucosa damage and multiple bowel diseases. Hydroxyproline (Hyp) is an imino acid abundant in sow's milk. Compelling evidence has been gathered showing the potential antioxidative properties of Hyp. However, the role and mechanism of Hyp in porcine intestinal epithelial cells in response to oxidative stress remains unknown. In this study, small intestinal epithelial cell lines of piglets (IPEC-1) were used to evaluate the protective effects of Hyp on 4-hydroxy-2-nonenal (4-HNE)-induced oxidative DNA damage and apoptosis. IPEC-1 pretreated with 0.5 to 5 mmol/L Hyp were exposed to 4-HNE (40 μmol/L) in the presence or absence of Hyp. Thereafter, the cells were subjected to apoptosis detection by Hoechst staining, flow cytometry, and Western blot or DNA damage analysis by comet assay, immunofluorescence, and reverse-transcription quantitative PCR (RT-qPCR). Cell apoptosis and the upregulation of cleaved-caspase-3 induced by 4-HNE (40 μmol/L) were inhibited by 5 mmol/L of Hyp. In addition, 5 mmol/L Hyp attenuated 4-HNE-induced reactive oxygen species (ROS) accumulation, glutathione (GSH) deprivation and DNA damage. The elevation in transcription of GADD45a (growth arrest and DNA-damage-inducible protein 45 alpha) and GADD45b (growth arrest and DNA-damage-inducible protein 45 beta), as well as the phosphorylation of H2AX (H2A histone family, member X), p38 MAPK (mitogen-activated protein kinase), and JNK (c-Jun N-terminal kinase) in cells treated with 4-HNE were alleviated by 5 mmol/L Hyp. Furthermore, Hyp supplementation increased the protein abundance of Krüppel like factor 4 (KLF4) in cells exposed to 4-HNE. Suppression of KLF4 expression by kenpaulone impeded the resistance of Hyp-treated cells to DNA damage and apoptosis induced by 4-HNE. Collectively, our results indicated that Hyp serves to protect against 4-HNE-induced apoptosis and DNA damage in IPEC-1 cells, which is partially pertinent with the enhanced expression of KLF4. Our data provides an updated explanation for the nutritional values of Hyp-containing animal products.
Collapse
Affiliation(s)
- Yun Ji
- State Key Laboratory of Animal Nutrition, Department of Animal Nutrition and Feed Science, China Agricultural University, Beijing 100193, China
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing 100193, China
| | - Yu He
- State Key Laboratory of Animal Nutrition, Department of Animal Nutrition and Feed Science, China Agricultural University, Beijing 100193, China
| | - Ying Yang
- State Key Laboratory of Animal Nutrition, Department of Animal Nutrition and Feed Science, China Agricultural University, Beijing 100193, China
| | - Zhaolai Dai
- State Key Laboratory of Animal Nutrition, Department of Animal Nutrition and Feed Science, China Agricultural University, Beijing 100193, China
| | - Zhenlong Wu
- State Key Laboratory of Animal Nutrition, Department of Animal Nutrition and Feed Science, China Agricultural University, Beijing 100193, China
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing 100193, China
- Corresponding author.
| |
Collapse
|
23
|
Thomas AF, Kelly GL, Strasser A. Of the many cellular responses activated by TP53, which ones are critical for tumour suppression? Cell Death Differ 2022; 29:961-971. [PMID: 35396345 PMCID: PMC9090748 DOI: 10.1038/s41418-022-00996-z] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 03/24/2022] [Accepted: 03/28/2022] [Indexed: 12/12/2022] Open
Abstract
The tumour suppressor TP53 is a master regulator of several cellular processes that collectively suppress tumorigenesis. The TP53 gene is mutated in ~50% of human cancers and these defects usually confer poor responses to therapy. The TP53 protein functions as a homo-tetrameric transcription factor, directly regulating the expression of ~500 target genes, some of them involved in cell death, cell cycling, cell senescence, DNA repair and metabolism. Originally, it was thought that the induction of apoptotic cell death was the principal mechanism by which TP53 prevents the development of tumours. However, gene targeted mice lacking the critical effectors of TP53-induced apoptosis (PUMA and NOXA) do not spontaneously develop tumours. Indeed, even mice lacking the critical mediators for TP53-induced apoptosis, G1/S cell cycle arrest and cell senescence, namely PUMA, NOXA and p21, do not spontaneously develop tumours. This suggests that TP53 must activate additional cellular responses to mediate tumour suppression. In this review, we will discuss the processes by which TP53 regulates cell death, cell cycling/cell senescence, DNA damage repair and metabolic adaptation, and place this in context of current understanding of TP53-mediated tumour suppression.
Collapse
Affiliation(s)
- Annabella F Thomas
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia.,The Department of Medical Biology, The University of Melbourne, Melbourne, VIC, Australia
| | - Gemma L Kelly
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia.,The Department of Medical Biology, The University of Melbourne, Melbourne, VIC, Australia
| | - Andreas Strasser
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia. .,The Department of Medical Biology, The University of Melbourne, Melbourne, VIC, Australia.
| |
Collapse
|
24
|
Li J, Long J, Zhang J, Liu N, Yan B, Tang L, Chen X, Peng C. Novel chloroquine derivative suppresses melanoma cell growth by DNA damage through increasing ROS levels. J Cell Mol Med 2022; 26:2579-2593. [PMID: 35332658 PMCID: PMC9077290 DOI: 10.1111/jcmm.17260] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 02/18/2022] [Accepted: 02/21/2022] [Indexed: 11/30/2022] Open
Abstract
Melanoma is a fatal cancer with a significant feature of resistance to traditional chemotherapeutic drugs and radiotherapy. A mutation in the kinase BRAF is observed in more than 66% of metastatic melanoma cases. Therefore, there is an urgent need to develop new BRAF-mutant melanoma inhibitors. High-dose chloroquine has been reported to have antitumour effects, but it often induces dose-limiting toxicity. In this study, a series of chloroquine derivatives were synthesized, and lj-2-66 had the best activity and was selected for further investigation. Furthermore, the anti-BRAF-mutant melanoma effect and mechanism of this compound were explored. CCK-8 and colony formation assays indicated that lj-2-66 significantly inhibited the proliferation of BRAF-mutant melanoma cells. Flow cytometry revealed that lj-2-66 induced G2/M arrest in melanoma cells and promoted apoptosis. Furthermore, lj-2-66 increased the level of ROS in melanoma cells and induced DNA damage. Interestingly, lj-2-66 also played a similar role in BRAF inhibitor-resistant melanoma cells. In summary, we found a novel chloroquine derivative, lj-2-66, that increased the level of ROS in melanoma cells and induced DNA damage, thus leading to G2/M arrest and apoptosis. These findings indicated that lj-2-66 may become a potential therapeutic drug for melanoma harbouring BRAF mutations.
Collapse
Affiliation(s)
- Jiaoduan Li
- Department of DermatologyXiangya Hospital, Central South UniversityChangshaChina
- Hunan Key Laboratory of Skin Cancer and PsoriasisXiangya HospitalChangshaChina
- Hunan Engineering Research Center of Skin Health and DiseaseXiangya HospitalChangshaChina
- National Clinical Research Center for Geriatric DisordersXiangya Hospital, Central South UniversityChangshaChina
| | - Jing Long
- Department of DermatologyXiangya Hospital, Central South UniversityChangshaChina
- Hunan Key Laboratory of Skin Cancer and PsoriasisXiangya HospitalChangshaChina
- Hunan Engineering Research Center of Skin Health and DiseaseXiangya HospitalChangshaChina
- National Clinical Research Center for Geriatric DisordersXiangya Hospital, Central South UniversityChangshaChina
| | - Jianglin Zhang
- Department of Dermatology, Shenzhen People’s Hospital, The Second Clinical Medical College, Jinan University, The First Affiliated HospitalSouthern University of Science and TechnologyShenzhenChina
| | - Nian Liu
- Department of DermatologyXiangya Hospital, Central South UniversityChangshaChina
- Hunan Key Laboratory of Skin Cancer and PsoriasisXiangya HospitalChangshaChina
- Hunan Engineering Research Center of Skin Health and DiseaseXiangya HospitalChangshaChina
- National Clinical Research Center for Geriatric DisordersXiangya Hospital, Central South UniversityChangshaChina
| | - Bei Yan
- Department of DermatologyXiangya Hospital, Central South UniversityChangshaChina
- Hunan Key Laboratory of Skin Cancer and PsoriasisXiangya HospitalChangshaChina
- Hunan Engineering Research Center of Skin Health and DiseaseXiangya HospitalChangshaChina
- National Clinical Research Center for Geriatric DisordersXiangya Hospital, Central South UniversityChangshaChina
| | - Ling Tang
- Department of DermatologyXiangya Hospital, Central South UniversityChangshaChina
- Hunan Key Laboratory of Skin Cancer and PsoriasisXiangya HospitalChangshaChina
- Hunan Engineering Research Center of Skin Health and DiseaseXiangya HospitalChangshaChina
- National Clinical Research Center for Geriatric DisordersXiangya Hospital, Central South UniversityChangshaChina
| | - Xiang Chen
- Department of DermatologyXiangya Hospital, Central South UniversityChangshaChina
- Hunan Key Laboratory of Skin Cancer and PsoriasisXiangya HospitalChangshaChina
- Hunan Engineering Research Center of Skin Health and DiseaseXiangya HospitalChangshaChina
- National Clinical Research Center for Geriatric DisordersXiangya Hospital, Central South UniversityChangshaChina
| | - Cong Peng
- Department of DermatologyXiangya Hospital, Central South UniversityChangshaChina
- Hunan Key Laboratory of Skin Cancer and PsoriasisXiangya HospitalChangshaChina
- Hunan Engineering Research Center of Skin Health and DiseaseXiangya HospitalChangshaChina
- National Clinical Research Center for Geriatric DisordersXiangya Hospital, Central South UniversityChangshaChina
| |
Collapse
|
25
|
Cona B, Hayashi T, Yamada A, Shimizu N, Yokota N, Nakato R, Shirahige K, Akiyama T. The splicing factor DHX38/PRP16 is required for ovarian clear cell carcinoma tumorigenesis, as revealed by a CRISPR-Cas9 screen. FEBS Open Bio 2021; 12:582-593. [PMID: 34965029 PMCID: PMC8886329 DOI: 10.1002/2211-5463.13358] [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: 09/02/2021] [Revised: 11/20/2021] [Accepted: 12/28/2021] [Indexed: 11/30/2022] Open
Abstract
Certain cancers, such as ovarian clear cell carcinoma (OCCC), display high levels of genetic variation between patients, making it difficult to develop effective therapies. In order to identify novel genes critical to OCCC growth, we carried out a comprehensive CRISPR‐Cas9 knockout screen against cell growth using an OCCC cell line and a normal ovarian surface epithelium cell line. We identified the gene encoding DHX38/PRP16, an ATP‐dependent RNA helicase involved in splicing, as critical for the growth and tumorigenesis of OCCC. DHX38/PRP16 knockdown in OCCC cells, but not normal cells, induces apoptosis and impairs OCCC tumorigenesis in a mouse model. Our results suggest that DHX38/PRP16 may play a role in OCCC tumorigenesis and could potentially be a promising therapeutic target.
Collapse
Affiliation(s)
- Brandon Cona
- Laboratory of Molecular and Genetic Information, Institute of Quantitative Biosciences, The University of Tokyo, 1-1-1 Yayoi Bunkyo-ku, Tokyo, 113-0032, Japan
| | - Tomoatsu Hayashi
- Laboratory of Molecular and Genetic Information, Institute of Quantitative Biosciences, The University of Tokyo, 1-1-1 Yayoi Bunkyo-ku, Tokyo, 113-0032, Japan
| | - Ai Yamada
- Laboratory of Molecular and Genetic Information, Institute of Quantitative Biosciences, The University of Tokyo, 1-1-1 Yayoi Bunkyo-ku, Tokyo, 113-0032, Japan
| | - Naomi Shimizu
- Laboratory of Molecular and Genetic Information, Institute of Quantitative Biosciences, The University of Tokyo, 1-1-1 Yayoi Bunkyo-ku, Tokyo, 113-0032, Japan
| | - Naoko Yokota
- Laboratory of Computational Genetics, Institute of Quantitative Biosciences, The University of Tokyo, 1-1-1 Yayoi Bunkyo-ku, Tokyo, 113-0032, Japan
| | - Ryuichiro Nakato
- Laboratory of Computational Genetics, Institute of Quantitative Biosciences, The University of Tokyo, 1-1-1 Yayoi Bunkyo-ku, Tokyo, 113-0032, Japan
| | - Katsuhiko Shirahige
- Laboratory of Genome Structure and Function, Institute of Quantitative Biosciences, The University of Tokyo, 1-1-1 Yayoi Bunkyo-ku, Tokyo, 113-0032, Japan
| | - Tetsu Akiyama
- Laboratory of Molecular and Genetic Information, Institute of Quantitative Biosciences, The University of Tokyo, 1-1-1 Yayoi Bunkyo-ku, Tokyo, 113-0032, Japan
| |
Collapse
|
26
|
Elersek T, Novak M, Mlinar M, Virant I, Bahor N, Leben K, Žegura B, Filipič M. Lethal and Sub-Lethal Effects and Modulation of Gene Expression Induced by T Kinase Inhibitors in Zebrafish (Danio Rerio) Embryos. TOXICS 2021; 10:toxics10010004. [PMID: 35051046 PMCID: PMC8781212 DOI: 10.3390/toxics10010004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 12/16/2021] [Accepted: 12/21/2021] [Indexed: 12/11/2022]
Abstract
Tyrosine kinase inhibitors (TKIs) are designed for targeted cancer therapy. The consumption of these drugs during the last 20 years has been constantly rising. In the zebrafish (Danio rerio) embryo toxicity test, we assessed the toxicity of six TKIs: imatinib mesylate, erlotinib, nilotinib, dasatinib, sorafenib and regorafenib. Imatinib mesylate and dasatinib induced lethal effects, while regorafenib, sorfenib and dasatinib caused a significant increase of sub-lethal effects, predominantly oedema, no blood circulation and formation of blood aggregates. The analyses of the changes in the expression of selected genes associated with the hormone system after the exposure to imatinib mesylate, dasatinib and regorafenib demonstrated that all three tested TKIs deregulated the expression of oestrogen receptor esr1, cytochrome P450 aromatase (cypa19b) and hydroxysteroid-dehydrogenase (hsd3b), regorafenib, and also thyroglobulin (tg). The expression of genes involved in the DNA damage response (gadd45 and mcm6) and apoptosis (bcl2) was deregulated only by exposure to regorafenib. The data indicate that common mechanisms, namely antiangiogenic activity and interference with steroidogenesis are involved in the TKI induced sub-lethal effects and potential hormone disrupting activity, respectively. The residues of TKIs may represent an environmental hazard; therefore, further ecotoxicological studies focusing also on the effects of their mixtures are warranted.
Collapse
Affiliation(s)
- Tina Elersek
- Department of Genetic Toxicology and Cancer Biology, National Institute of Biology, Večna pot 111, 1000 Ljubljana, Slovenia; (T.E.); (M.N.); (M.M.); (N.B.); (K.L.); (B.Ž.)
| | - Matjaž Novak
- Department of Genetic Toxicology and Cancer Biology, National Institute of Biology, Večna pot 111, 1000 Ljubljana, Slovenia; (T.E.); (M.N.); (M.M.); (N.B.); (K.L.); (B.Ž.)
| | - Mateja Mlinar
- Department of Genetic Toxicology and Cancer Biology, National Institute of Biology, Večna pot 111, 1000 Ljubljana, Slovenia; (T.E.); (M.N.); (M.M.); (N.B.); (K.L.); (B.Ž.)
| | - Igor Virant
- Institute of Oncology Ljubljana, Zaloška 2, 1000 Ljubljana, Slovenia;
| | - Nika Bahor
- Department of Genetic Toxicology and Cancer Biology, National Institute of Biology, Večna pot 111, 1000 Ljubljana, Slovenia; (T.E.); (M.N.); (M.M.); (N.B.); (K.L.); (B.Ž.)
- Biotechnical Faculty, University of Ljubljana, Jamnikarjeva 101, 1000 Ljubljana, Slovenia
| | - Karin Leben
- Department of Genetic Toxicology and Cancer Biology, National Institute of Biology, Večna pot 111, 1000 Ljubljana, Slovenia; (T.E.); (M.N.); (M.M.); (N.B.); (K.L.); (B.Ž.)
- Biotechnical Faculty, University of Ljubljana, Jamnikarjeva 101, 1000 Ljubljana, Slovenia
| | - Bojana Žegura
- Department of Genetic Toxicology and Cancer Biology, National Institute of Biology, Večna pot 111, 1000 Ljubljana, Slovenia; (T.E.); (M.N.); (M.M.); (N.B.); (K.L.); (B.Ž.)
| | - Metka Filipič
- Department of Genetic Toxicology and Cancer Biology, National Institute of Biology, Večna pot 111, 1000 Ljubljana, Slovenia; (T.E.); (M.N.); (M.M.); (N.B.); (K.L.); (B.Ž.)
- Correspondence:
| |
Collapse
|
27
|
Rizzotto D, Englmaier L, Villunger A. At a Crossroads to Cancer: How p53-Induced Cell Fate Decisions Secure Genome Integrity. Int J Mol Sci 2021; 22:ijms221910883. [PMID: 34639222 PMCID: PMC8509445 DOI: 10.3390/ijms221910883] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 09/29/2021] [Accepted: 10/01/2021] [Indexed: 12/12/2022] Open
Abstract
P53 is known as the most critical tumor suppressor and is often referred to as the guardian of our genome. More than 40 years after its discovery, we are still struggling to understand all molecular details on how this transcription factor prevents oncogenesis or how to leverage current knowledge about its function to improve cancer treatment. Multiple cues, including DNA-damage or mitotic errors, can lead to the stabilization and nuclear translocation of p53, initiating the expression of multiple target genes. These transcriptional programs may be cell-type- and stimulus-specific, as is their outcome that ultimately imposes a barrier to cellular transformation. Cell cycle arrest and cell death are two well-studied consequences of p53 activation, but, while being considered critical, they do not fully explain the consequences of p53 loss-of-function phenotypes in cancer. Here, we discuss how mitotic errors alert the p53 network and give an overview of multiple ways that p53 can trigger cell death. We argue that a comparative analysis of different types of p53 responses, elicited by different triggers in a time-resolved manner in well-defined model systems, is critical to understand the cell-type-specific cell fate induced by p53 upon its activation in order to resolve the remaining mystery of its tumor-suppressive function.
Collapse
Affiliation(s)
- Dario Rizzotto
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1090 Vienna, Austria; (D.R.); (L.E.)
| | - Lukas Englmaier
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1090 Vienna, Austria; (D.R.); (L.E.)
- Ludwig Boltzmann Institute for Rare and Undiagnosed Diseases (LBI-RUD), 1090 Vienna, Austria
| | - Andreas Villunger
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1090 Vienna, Austria; (D.R.); (L.E.)
- Ludwig Boltzmann Institute for Rare and Undiagnosed Diseases (LBI-RUD), 1090 Vienna, Austria
- Institute for Developmental Immunology, Biocenter, Medical University of Innsbruck, 6020 Innsbruck, Austria
- Correspondence:
| |
Collapse
|
28
|
Raghunandan S, Ramachandran S, Ke E, Miao Y, Lal R, Chen ZB, Subramaniam S. Heme Oxygenase-1 at the Nexus of Endothelial Cell Fate Decision Under Oxidative Stress. Front Cell Dev Biol 2021; 9:702974. [PMID: 34595164 PMCID: PMC8476872 DOI: 10.3389/fcell.2021.702974] [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: 04/30/2021] [Accepted: 08/17/2021] [Indexed: 12/31/2022] Open
Abstract
Endothelial cells (ECs) form the inner lining of blood vessels and are central to sensing chemical perturbations that can lead to oxidative stress. The degree of stress is correlated with divergent phenotypes such as quiescence, cell death, or senescence. Each possible cell fate is relevant for a different aspect of endothelial function, and hence, the regulation of cell fate decisions is critically important in maintaining vascular health. This study examined the oxidative stress response (OSR) in human ECs at the boundary of cell survival and death through longitudinal measurements, including cellular, gene expression, and perturbation measurements. 0.5 mM hydrogen peroxide (HP) produced significant oxidative stress, placed the cell at this junction, and provided a model to study the effectors of cell fate. The use of systematic perturbations and high-throughput measurements provide insights into multiple regimes of the stress response. Using a systems approach, we decipher molecular mechanisms across these regimes. Significantly, our study shows that heme oxygenase-1 (HMOX1) acts as a gatekeeper of cell fate decisions. Specifically, HP treatment of HMOX1 knockdown cells reversed the gene expression of about 51% of 2,892 differentially expressed genes when treated with HP alone, affecting a variety of cellular processes, including anti-oxidant response, inflammation, DNA injury and repair, cell cycle and growth, mitochondrial stress, metabolic stress, and autophagy. Further analysis revealed that these switched genes were highly enriched in three spatial locations viz., cell surface, mitochondria, and nucleus. In particular, it revealed the novel roles of HMOX1 on cell surface receptors EGFR and IGFR, mitochondrial ETCs (MTND3, MTATP6), and epigenetic regulation through chromatin modifiers (KDM6A, RBBP5, and PPM1D) and long non-coding RNA (lncRNAs) in orchestrating the cell fate at the boundary of cell survival and death. These novel aspects suggest that HMOX1 can influence transcriptional and epigenetic modulations to orchestrate OSR affecting cell fate decisions.
Collapse
Affiliation(s)
- Sindhushree Raghunandan
- Department of Bioengineering, University of California, San Diego, San Diego, CA, United States
| | - Srinivasan Ramachandran
- Department of Bioengineering, University of California, San Diego, San Diego, CA, United States
| | - Eugene Ke
- Department of Bioengineering, University of California, San Diego, San Diego, CA, United States
| | - Yifei Miao
- Department of Diabetes Complications and Metabolism, City of Hope, Duarte, CA, United States
| | - Ratnesh Lal
- Department of Bioengineering, University of California, San Diego, San Diego, CA, United States.,Department of Mechanical and Aerospace Engineering, University of California, San Diego, San Diego, CA, United States
| | - Zhen Bouman Chen
- Department of Diabetes Complications and Metabolism, City of Hope, Duarte, CA, United States
| | - Shankar Subramaniam
- Department of Bioengineering, University of California, San Diego, San Diego, CA, United States.,Department of Computer Science and Engineering, University of California, San Diego, San Diego, CA, United States.,Department of Cellular and Molecular Medicine, University of California, San Diego, San Diego, CA, United States
| |
Collapse
|
29
|
Del Corvo M, Lazzari B, Capra E, Zavarez L, Milanesi M, Utsunomiya YT, Utsunomiya ATH, Stella A, de Paula Nogueira G, Garcia JF, Ajmone-Marsan P. Methylome Patterns of Cattle Adaptation to Heat Stress. Front Genet 2021; 12:633132. [PMID: 34122501 PMCID: PMC8194315 DOI: 10.3389/fgene.2021.633132] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 05/04/2021] [Indexed: 12/13/2022] Open
Abstract
Heat stress has a detrimental impact on cattle health, welfare and productivity by affecting gene expression, metabolism and immune response, but little is known on the epigenetic mechanisms mediating the effect of temperature at the cellular and organism level. In this study, we investigated genome-wide DNA methylation in blood samples collected from 5 bulls of the heat stress resilient Nellore breed and 5 bulls of the Angus that are more heat stress susceptible, exposed to the sun and high temperature-high humidity during the summer season of the Brazilian South-East region. The methylomes were analyzed during and after the exposure by Reduced Representation Bisulfite Sequencing, which provided genome-wide single-base resolution methylation profiles. Significant methylation changes between stressful and recovery periods were observed in 819 genes. Among these, 351 were only seen in Angus, 366 were specific to Nellore, and 102 showed significant changes in methylation patterns in both breeds. KEGG and Gene Ontology (GO) enrichment analyses showed that responses were breed-specific. Interestingly, in Nellore significant genes and pathways were mainly involved in stress responses and cellular defense and were under methylated during heat stress, whereas in Angus the response was less focused. These preliminary results suggest that heat challenge induces changes in methylation patterns in specific loci, which should be further scrutinized to assess their role in heat tolerance.
Collapse
Affiliation(s)
- Marcello Del Corvo
- Department of Animal Science Food and Nutrition - DIANA, Nutrigenomics and Proteomics Research Centre - PRONUTRIGEN, and Biodiversity and Ancient DNA Research Centre, Università Cattolica del Sacro Cuore, Piacenza, Italy.,Istituto di Biologia e Biotecnologia Agraria, Consiglio Nazionale delle Ricerche IBBA CNR, Milan, Italy
| | - Barbara Lazzari
- Istituto di Biologia e Biotecnologia Agraria, Consiglio Nazionale delle Ricerche IBBA CNR, Milan, Italy
| | - Emanuele Capra
- Istituto di Biologia e Biotecnologia Agraria, Consiglio Nazionale delle Ricerche IBBA CNR, Milan, Italy
| | - Ludmilla Zavarez
- School of Veterinary Medicine, Araçatuba, Department of Production and Animal Health, São Paulo State University (unesp), Araçatuba, Brazil.,International Atomic Energy Agency, Collaborating Centre on Animal Genomics and Bioinformatics, Araçatuba, Brazil
| | - Marco Milanesi
- School of Veterinary Medicine, Araçatuba, Department of Production and Animal Health, São Paulo State University (unesp), Araçatuba, Brazil.,International Atomic Energy Agency, Collaborating Centre on Animal Genomics and Bioinformatics, Araçatuba, Brazil
| | - Yuri Tani Utsunomiya
- School of Veterinary Medicine, Araçatuba, Department of Production and Animal Health, São Paulo State University (unesp), Araçatuba, Brazil.,International Atomic Energy Agency, Collaborating Centre on Animal Genomics and Bioinformatics, Araçatuba, Brazil
| | - Adam Taiti Harth Utsunomiya
- School of Veterinary Medicine, Araçatuba, Department of Production and Animal Health, São Paulo State University (unesp), Araçatuba, Brazil.,International Atomic Energy Agency, Collaborating Centre on Animal Genomics and Bioinformatics, Araçatuba, Brazil
| | - Alessandra Stella
- Istituto di Biologia e Biotecnologia Agraria, Consiglio Nazionale delle Ricerche IBBA CNR, Milan, Italy
| | - Guilherme de Paula Nogueira
- School of Veterinary Medicine, Araçatuba, Department of Production and Animal Health, São Paulo State University (unesp), Araçatuba, Brazil
| | - Josè Fernando Garcia
- School of Veterinary Medicine, Araçatuba, Department of Production and Animal Health, São Paulo State University (unesp), Araçatuba, Brazil.,International Atomic Energy Agency, Collaborating Centre on Animal Genomics and Bioinformatics, Araçatuba, Brazil
| | - Paolo Ajmone-Marsan
- Department of Animal Science Food and Nutrition - DIANA, Nutrigenomics and Proteomics Research Centre - PRONUTRIGEN, and Biodiversity and Ancient DNA Research Centre, Università Cattolica del Sacro Cuore, Piacenza, Italy
| |
Collapse
|
30
|
Yang Y, Shang H. Silencing lncRNA-DGCR5 increased trophoblast cell migration, invasion and tube formation, and inhibited cell apoptosis via targeting miR-454-3p/GADD45A axis. Mol Cell Biochem 2021; 476:3407-3421. [PMID: 33973132 DOI: 10.1007/s11010-021-04161-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 04/15/2021] [Indexed: 12/26/2022]
Abstract
Long noncoding RNA (lncRNA)-DGCR5 has been recognized as a potential tumor progression regulator, while its expression and specific functions in preeclampsia (PE) development remain unveiled. The expressions of miR-454-3p, lncRNA-DiGeorge syndrome critical region gene 5 (DGCR5) and growth arrest and DNA damage protein-inducible 45A (GADD45A) in placental tissues from PE patients or HTR-8/SVneo cells were assessed by Western blot or qRT-PCR. Dual-luciferase reporter assay determined the binding relations between miR-454-3p and GADD45A and between miR-454-3p and lncRNA-DGCR5. The viability, apoptosis, migration, invasiveness and tube formation of HTR-8/SVneo cell were evaluated using cell counting kit (CCK)-8, Annexin-V/Propidium iodide staining, wound healing, transwell and tube formation assays, respectively. miR-454-3p was low-expressed in PE tissue, and upregulation of miR-454-3p increased viability and promoted migration, invasion and tube formation in HTR-8/SVneo cells while inhibiting apoptosis. Then, miR-454-3p was found to directly target GADD45A which was high-expressed in PE tissues. Overexpressing GADD45A decreased the viability and inhibited the migration, invasion and tube formation of HTR-8/SVneo cells while enhancing apoptosis, and it neutralized the effect of miR-454-3p upregulation. In turn, miR-454-3p upregulation reversed the effect of GADD45A overexpression. Meanwhile, miR-454-3p could also target lncRNA-DGCR5. Silencing lncRNA-DGCR5 increased miR-454-3p expression and cell viability and promoted migration, invasion and tube formation in HTR-8/SVneo cells while inhibiting apoptosis, and it counteracted the effect of miR-454-3p downregulation. As usual, miR-454-3p downregulation reversed the effect of lncRNA-DGCR5 silencing. To conclude, silencing lncRNA-DGCR5 increased viability, promoted migration, invasion and tube formation, and inhibited apoptosis in HTR-8/SVneo cells by rescuing the inhibition of GADD45A expression caused by miR-454-3p.
Collapse
Affiliation(s)
- Yanlin Yang
- Department of Obstetrics and Gynecology, Shanxi Bethune Hospital Shanxi Academy of Medical Sciences, No.99, Longcheng Street, Taiyuan, 030032, China.
| | - Haixia Shang
- Department of Obstetrics and Gynecology, Shanxi Bethune Hospital Shanxi Academy of Medical Sciences, No.99, Longcheng Street, Taiyuan, 030032, China
| |
Collapse
|
31
|
Transcriptomic Changes Associated with Loss of Cell Viability Induced by Oxysterol Treatment of a Retinal Photoreceptor-Derived Cell Line: An In Vitro Model of Smith-Lemli-Opitz Syndrome. Int J Mol Sci 2021; 22:ijms22052339. [PMID: 33652836 PMCID: PMC7956713 DOI: 10.3390/ijms22052339] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 02/19/2021] [Accepted: 02/21/2021] [Indexed: 11/17/2022] Open
Abstract
Smith–Lemli–Opitz Syndrome (SLOS) results from mutations in the gene encoding the enzyme DHCR7, which catalyzes conversion of 7-dehydrocholesterol (7DHC) to cholesterol (CHOL). Rats treated with a DHCR7 inhibitor serve as a SLOS animal model, and exhibit progressive photoreceptor-specific cell death, with accumulation of 7DHC and oxidized sterols. To understand the basis of this cell type specificity, we performed transcriptomic analyses on a photoreceptor-derived cell line (661W), treating cells with two 7DHC-derived oxysterols, which accumulate in tissues and bodily fluids of SLOS patients and in the rat SLOS model, as well as with CHOL (negative control), and evaluated differentially expressed genes (DEGs) for each treatment. Gene enrichment analysis and compilation of DEG sets indicated that endoplasmic reticulum stress, oxidative stress, DNA damage and repair, and autophagy were all highly up-regulated pathways in oxysterol-treated cells. Detailed analysis indicated that the two oxysterols exert their effects via different molecular mechanisms. Changes in expression of key genes in highlighted pathways (Hmox1, Ddit3, Trib3, and Herpud1) were validated by immunofluorescence confocal microscopy. The results extend our understanding of the pathobiology of retinal degeneration and SLOS, identifying potential new druggable targets for therapeutic intervention into these and other related orphan diseases.
Collapse
|
32
|
tRNA Lys-Derived Fragment Alleviates Cisplatin-Induced Apoptosis in Prostate Cancer Cells. Pharmaceutics 2021; 13:pharmaceutics13010055. [PMID: 33406670 PMCID: PMC7824007 DOI: 10.3390/pharmaceutics13010055] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 12/29/2020] [Accepted: 12/30/2020] [Indexed: 02/06/2023] Open
Abstract
Cisplatin is a standard treatment for prostate cancer, which is the third leading cause of cancer-related deaths among men globally. However, patients who have undergone cisplatin can rxperience relapse. tRNA-derived fragments (tRFs) are small non-coding RNAs generated via tRNA cleavage; their physiological activities are linked to the development of human diseases. Specific tRFs, including tRF-315 derived from tRNALys, are highly expressed in prostate cancer patients. However, whether tRF-315 regulates prostate cancer cell proliferation or apoptosis is unclear. Herein, we confirmed that tRF-315 expression was higher in prostate cancer cells (LNCaP, DU145, and PC3) than in normal prostate cells. tRF-315 prevented cisplatin-induced apoptosis and alleviated cisplatin-induced mitochondrial dysfunction in LNCaP and DU145 cells. Moreover, transfection of tRF-315 inhibitor increased the expression of apoptotic pathway-related proteins in LNCaP and DU145 cells. Furthermore, tRF-315 targeted the tumor suppressor gene GADD45A, thus regulating the cell cycle, which was altered by cisplatin in LNCaP and DU145 cells. Thus, tRF-315 protects prostate cancer cells from mitochondrion-dependent apoptosis induced by cisplatin treatment.
Collapse
|
33
|
Chen G, Wang L, Wang M, Hu T. Comprehensive insights into the occurrence and toxicological issues of nodularins. MARINE POLLUTION BULLETIN 2021; 162:111884. [PMID: 33307402 DOI: 10.1016/j.marpolbul.2020.111884] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 11/01/2020] [Accepted: 11/23/2020] [Indexed: 06/12/2023]
Abstract
The occurrence of cyanobacterial toxins is being increasingly reported. Nodularins (NODs) are one of the cyanotoxins group mainly produced by Nodularia spumigena throughout the world. NODs may exert adverse effects on animal and human health, and NOD-R variant is the most widely investigated. However, research focused on them is still limited. In order to understand the realistic risk well, the aim of this review is to compile the available information in the scientific literature regarding NODs, including their sources, distribution, structural characteristics, physicochemical properties, biosynthesis and degradation, adverse effects in vitro and vivo, and toxicokinetics. More data is urgently needed to integrate the cumulative or synergistic effects of NODs on different species and various cells to better understand, anticipate and aggressively manage their potential toxicity after both short- and long-term exposure in ecosystem, and to minimize or prevent the adverse effects on human health, environment and the economy.
Collapse
Affiliation(s)
- Guoliang Chen
- Key Laboratory of Biorheological Science and Technology (Chongqing University), Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400030, China
| | - Linping Wang
- Key Laboratory of Biorheological Science and Technology (Chongqing University), Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400030, China
| | - Mingxing Wang
- Key Laboratory of Biorheological Science and Technology (Chongqing University), Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400030, China
| | - Tingzhang Hu
- Key Laboratory of Biorheological Science and Technology (Chongqing University), Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400030, China.
| |
Collapse
|
34
|
Gene Expression Alterations Associated with Oleuropein-Induced Antiproliferative Effects and S-Phase Cell Cycle Arrest in Triple-Negative Breast Cancer Cells. Nutrients 2020; 12:nu12123755. [PMID: 33297339 PMCID: PMC7762327 DOI: 10.3390/nu12123755] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 11/18/2020] [Accepted: 12/04/2020] [Indexed: 02/06/2023] Open
Abstract
It is known that the Mediterranean diet is effective in reducing the risk of several chronic diseases, including cancer. A critical component of the Mediterranean diet is olive oil, and the relationship between olive oil consumption and the reduced risk of cancer has been established. Oleuropein (OL) is the most prominent polyphenol component of olive fruits and leaves. This compound has been shown to have potent properties in various types of cancers, including breast cancer. In the present study, the molecular mechanism of OL was examined in two racially different triple-negative breast cancer (TNBC) cell lines-African American (AA, MDA-MB-468) and Caucasian American (CA, MDA-MB-231). The data obtained showed that OL effectively inhibits cell growth in both cell lines, concomitant with S-phase cell cycle arrest-mediated apoptosis. The results also showed that OL-treated MDA-MB-468 cells were two-fold more sensitive to OL antiproliferative effect than MDA-MB-231 cells were. At lower concentrations, OL modified the expression of many apoptosis-involved genes. OL was more effective in MDA-MB-468, compared to MDA-MB-231 cells, in terms of the number and the fold-change of the altered genes. In MDA-MB-468 cells, OL induced a noticeable transcription activation in fourteen genes, including two members of the caspase family: caspase 1 (CASP1) and caspase 14 (CASP14); two members of the TNF receptor superfamily: Fas-associated via death domain (FADD) and TNF receptor superfamily 21 (TNFRSF21); six other proapoptotic genes: growth arrest and DNA damage-inducible 45 alpha (GADD45A), cytochrome c somatic (CYCS), BCL-2 interacting protein 2 (BNIP2), BCL-2 interacting protein 3 (BNIP3), BH3 interacting domain death agonist (BID), and B-cell lymphoma/leukemia 10 (BCL10); and the CASP8 and FADD-like apoptosis regulator (CFLAR) gene. Moreover, in MDA-MB-468 cells, OL induced a significant upregulation in two antiapoptotic genes: bifunctional apoptosis regulator (BFAR) and B-Raf proto-oncogene (BRAF) and a baculoviral inhibitor of apoptosis (IAP) repeat-containing 3 (BIRC3). On the contrary, in MDA-MB-231 cells, OL showed mixed impacts on gene expression. OL significantly upregulated the mRNA expression of four genes: BIRC3, receptor-interacting serine/threonine kinase 2 (RIPK2), TNF receptor superfamily 10A (TNFRSF10A), and caspase 4 (CASP4). Additionally, another four genes were repressed, including caspase 6 (CASP6), pyrin domain (PYD), and caspase recruitment domain (CARD)-containing (PAYCARD), baculoviral IAP repeat-containing 5 (BIRC5), and the most downregulated TNF receptor superfamily member 11B (TNFRSF11B, 16.34-fold). In conclusion, the data obtained indicate that the two cell lines were markedly different in the anticancer effect and mechanisms of oleuropein's ability to alter apoptosis-related gene expressions. The results obtained from this study should also guide the potential utilization of oleuropein as an adjunct therapy for TNBC to increase chemotherapy effectiveness and prevent cancer progression.
Collapse
|
35
|
Satta N, Weppe R, Pagano S, Frias M, Juillard C, Vuilleumier N. Auto-antibodies against apolipoprotein A-1 block cancer cells proliferation and induce apoptosis. Oncotarget 2020; 11:4266-4280. [PMID: 33245719 PMCID: PMC7679029 DOI: 10.18632/oncotarget.27814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Accepted: 10/27/2020] [Indexed: 11/25/2022] Open
Abstract
Auto-antibodies against apoA-1 (anti-apoA-1 IgGs) have been identified as important actors of atherosclerosis development through pro-inflammatory and pro-atherogenic properties and to also induce apoptosis in tumoral neuronal and lymphocyte derived cell lines through unknown mechanisms. The purpose of this study was to explore the cellular pathways involved in tumoral cell survival modulated by anti-apoA-1 antibodies. We observed that anti-apoA-1 antibodies induce growth arrest (in G2/M phase) and cell apoptosis through caspase 3 activation, accompanied by a selective p53 phosphorylation on serine 15. RNA sequencing indicated that anti-apoA-1 IgGs affect the expression of more than 950 genes belonging to five major groups of genes and respectively involved in i) cell proliferation inhibition, ii) p53 stabilisation and regulation, iii) apoptosis regulation, iv) inflammation regulation, and v) oxidative stress. In conclusion, anti-apoA-1 antibodies seem to have a role in blocking tumoral cell proliferation and survival, by activating a major tumor suppressor protein and by modulating the inflammatory and oxidative stress response. Further investigations are needed to explore a possible anti-cancer therapeutic approach of these antibodies in very specific and circumscribed conditions.
Collapse
Affiliation(s)
- Nathalie Satta
- Division of Laboratory Medicine, Department of Diagnostic, Geneva University Hospitals, Geneva, Switzerland.,Department of Medicine, Medical Faculty, Geneva University, Geneva, Switzerland
| | - Rémy Weppe
- Division of Laboratory Medicine, Department of Diagnostic, Geneva University Hospitals, Geneva, Switzerland.,Department of Medicine, Medical Faculty, Geneva University, Geneva, Switzerland
| | - Sabrina Pagano
- Division of Laboratory Medicine, Department of Diagnostic, Geneva University Hospitals, Geneva, Switzerland.,Department of Medicine, Medical Faculty, Geneva University, Geneva, Switzerland
| | - Miguel Frias
- Division of Laboratory Medicine, Department of Diagnostic, Geneva University Hospitals, Geneva, Switzerland.,Department of Medicine, Medical Faculty, Geneva University, Geneva, Switzerland
| | - Catherine Juillard
- Division of Laboratory Medicine, Department of Diagnostic, Geneva University Hospitals, Geneva, Switzerland.,Department of Medicine, Medical Faculty, Geneva University, Geneva, Switzerland
| | - Nicolas Vuilleumier
- Division of Laboratory Medicine, Department of Diagnostic, Geneva University Hospitals, Geneva, Switzerland.,Department of Medicine, Medical Faculty, Geneva University, Geneva, Switzerland
| |
Collapse
|
36
|
Mohammed ET, Hashem KS, Abdelazem AZ, Foda FAMA. Prospective Protective Effect of Ellagic Acid as a SIRT1 Activator in Iron Oxide Nanoparticle-Induced Renal Damage in Rats. Biol Trace Elem Res 2020; 198:177-188. [PMID: 31933277 DOI: 10.1007/s12011-020-02034-w] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 01/04/2020] [Indexed: 02/01/2023]
Abstract
Despite the wide application of iron oxide nanoparticles (IONPs), little is known about the specific mechanism of their nephrotoxic effect. We aimed to evaluate the nephrotoxic effects of iron oxide nanoparticles (IONPs) in vivo and the protective effect of ellagic acid (EA) as a silent information regulator sirtuin 1 (SIRT1) activator against the induced nephrotoxicity. Forty male albino Wistar rats were randomly distributed into four equal groups (10 rats each): the control group (oral saline for 30 days), ellagic acid (EA) group (10 mg/kg b.w. EA, orally for 30 days), IONP group (20 mg/kg b.w. IONP I/P injection at the 24th-30th day), and EA + IONP group (10 mg/kg b.w./day EA for 30 days + 20 mg/kg b.w. IONPs at the 24th-30th day). In the present study, the potent antioxidant and antiapoptotic effects of EA were indicated by the significant overexpression of SIRT1 in renal tissues that leads to significant decreases in renal MDA content, P53 protein level and forkhead-box transcription factor1 (FOXO1) expression, and significant increases in renal GSH level, catalase activity, growth arrest and DNA damage-inducible protein 45 alpha (GADDα45), and renal inhibition of apoptosis protein (KIAP) gene expression levels in the EA + IONP-treated group. These results were confirmed by the improved histopathological renal features with EA administration. In conclusion, the present study provides the first evidence for the usefulness of EA as a sirtuin1 activator in the prevention or treatment of renal damage. Thus, EA could be used as a promising therapy for the prevention of IONP-induced nephrotoxicity.
Collapse
Affiliation(s)
- Eman Taha Mohammed
- Department of Biochemistry, Faculty of Veterinary Medicine, Beni-Suef University, Beni-Suef, Egypt.
| | - Khalid S Hashem
- Department of Biochemistry, Faculty of Veterinary Medicine, Beni-Suef University, Beni-Suef, Egypt
| | - Ahmed Z Abdelazem
- Department of Biotechnology and Life Sciences, Faculty of Postgraduate Studies for Advanced Sciences, Beni-Suef University, Beni-Suef, Egypt
| | - Fatma A M A Foda
- Department of Biotechnology and Life Sciences, Faculty of Postgraduate Studies for Advanced Sciences, Beni-Suef University, Beni-Suef, Egypt
| |
Collapse
|
37
|
Messeha SS, Zarmouh NO, Asiri A, Soliman KFA. Rosmarinic acid-induced apoptosis and cell cycle arrest in triple-negative breast cancer cells. Eur J Pharmacol 2020; 885:173419. [PMID: 32750370 PMCID: PMC7541730 DOI: 10.1016/j.ejphar.2020.173419] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 07/23/2020] [Accepted: 07/24/2020] [Indexed: 01/06/2023]
Abstract
Rosmarinic acid (RA) is a polyphenolic compound with various pharmacological properties, including, anti-inflammatory, immunomodulatory, and neuroprotective, as well as having antioxidant and anticancer activities. This study evaluated the effects and mechanisms of RA in two racially different triple-negative breast cancer (TNBC) cell lines. Results obtained show that RA significantly caused cytotoxic and antiproliferative effects in both cell lines in a dose- and time-dependent manner. Remarkably, RA induced cell cycle arrest-related apoptosis and altered the expression of many apoptosis-involved genes differently. In MDA-MB-231 cells, RA arrested the cells in the G0/G1 phase. In contrast, the data suggest that RA causes S-phase arrest in MDA-MB-468 cells, leading to a 2-fold increase in the apoptotic effect compared to MDA-MB-231 cells. Further, in MDA-MB-231 cells, RA significantly upregulated the mRNA expression of three genes: harakiri (HRK), tumor necrosis factor receptor superfamily 25 (TNFRSF25), and BCL-2 interacting protein 3 (BNIP3). In contrast, in the MDA-MB-468 cell line, the compound induced a significant transcription activation in three genes, including TNF, growth arrest and DNA damage-inducible 45 alpha (GADD45A), and BNIP3. Furthermore, RA repressed the expression of TNF receptor superfamily 11B (TNFRSF11B) in MDA-MB-231 cells in comparison to the ligand TNF superfamily member 10 (TNFSF10) and baculoviral IAP repeat-containing 5 (BIRC5) in MDA-MB-468 cells. In conclusion, the data suggest that the polyphenol RA may have a potential role in TNBC therapies, particularly in MDA-MB-468 cells.
Collapse
Affiliation(s)
- Samia S Messeha
- Division of Pharmaceutical Sciences, College of Pharmacy & Pharmaceutical Sciences, Florida A&M University, 1415 ML King Blvd, Room G 134 H New Pharmacy Building, Tallahassee, FL, 32307, United States
| | - Najla O Zarmouh
- Division of Pharmaceutical Sciences, College of Pharmacy & Pharmaceutical Sciences, Florida A&M University, 1415 ML King Blvd, Room G 134 H New Pharmacy Building, Tallahassee, FL, 32307, United States
| | - Abrar Asiri
- Division of Pharmaceutical Sciences, College of Pharmacy & Pharmaceutical Sciences, Florida A&M University, 1415 ML King Blvd, Room G 134 H New Pharmacy Building, Tallahassee, FL, 32307, United States
| | - Karam F A Soliman
- Division of Pharmaceutical Sciences, College of Pharmacy & Pharmaceutical Sciences, Florida A&M University, 1415 ML King Blvd, Room G 134 H New Pharmacy Building, Tallahassee, FL, 32307, United States.
| |
Collapse
|
38
|
Najafzadeh B, Asadzadeh Z, Motafakker Azad R, Mokhtarzadeh A, Baghbanzadeh A, Alemohammad H, Abdoli Shadbad M, Vasefifar P, Najafi S, Baradaran B. The oncogenic potential of NANOG: An important cancer induction mediator. J Cell Physiol 2020; 236:2443-2458. [PMID: 32960465 DOI: 10.1002/jcp.30063] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 09/02/2020] [Accepted: 09/07/2020] [Indexed: 12/11/2022]
Abstract
Cancer stem cells (CSCs) are a unique population in the tumor, but they only comprise 2%-5% of the tumor bulk. Although CSCs share several features with embryonic stem cells, CSCs can give rise to the tumor cells. CSCs overexpress embryonic transcription factor NANOG, which is downregulated in differentiated tissues. This transcription factor confers CSC's stemness, unlimited self-renewal, metastasis, invasiveness, angiogenesis, and drug-resistance with the assistance of WNT, OCT4, SOX2, Hedgehog, BMI-1, and other complexes. NANOG facilitates CSCs development via multiple pathways, like angiogenesis and lessening E-cadherin expression levels, which paves the road for metastasis. Moreover, NANOG represses apoptosis and leads to drug-resistance. This review aims to highlight the pivotal role of NANOG and the pertained pathways in CSCs. Also, this current study intends to demonstrate that targeting NANOG can dimmish the CSCs, sensitize the tumor to chemotherapy, and eradicate the cancer cells.
Collapse
Affiliation(s)
- Basira Najafzadeh
- Department of Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| | - Zahra Asadzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Ahad Mokhtarzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amir Baghbanzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hajar Alemohammad
- Department of Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| | | | - Parisa Vasefifar
- Department of Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| | - Souzan Najafi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
39
|
Keratinocytes from Gorlin Syndrome-induced pluripotent stem cells are resistant against UV radiation. Med Mol Morphol 2020; 54:69-78. [PMID: 32816116 DOI: 10.1007/s00795-020-00264-4] [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: 06/01/2020] [Accepted: 07/28/2020] [Indexed: 10/23/2022]
Abstract
Gorlin syndrome (GS) is an autosomal dominant genetic disorder involving Patched 1 (PTCH1) mutations. The PTCH1 is a receptor as well as an inhibitor of hedgehog (Hh) to sequester downstream Hh pathway molecules called Smoothened (SMO). PTCH1 mutations causes a variety of GS conditions including falx calcification, odontogenic keratocytes and basal cell carcinomas (BCC). Because PTCH1 is a major driver gene of sporadic BCC, GS patients are characteristically prone to BCC. In order to elucidate the pathological mechanism of BCC-prone GS patients, we investigated keratinocytes derived from GS patient specific iPS cells (G-OFiPSCs) which were generated and reported previously. We found that keratinocytes derived from G-OFiPSCs (GKCs) have increased expression of Hh target molecules. GKCs were irradiated and those cells showed high resistance to UV induced apoptosis. BCL2, known as anti-apoptotic molecule as well as Hh target, significantly increased in GKCs. Several molecules involved in DNA repair, cell cycle control, senescence, and genotoxic stress such as TP53, BRCA1 and GADD45A increased only in GKCs. GKCs are indicated to be resistant to UV irradiation by upregulating molecules which control DNA repair and genotoxic even under DNA damage caused by UV. The anti-apoptotic properties of GKCs may contribute BCC.
Collapse
|
40
|
Angeles AK, Heckmann D, Flosdorf N, Duensing S, Sültmann H. The ERG-Regulated LINC00920 Promotes Prostate Cancer Cell Survival via the 14-3-3ε-FOXO Pathway. Mol Cancer Res 2020; 18:1545-1559. [PMID: 32646965 DOI: 10.1158/1541-7786.mcr-20-0021] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 06/04/2020] [Accepted: 06/29/2020] [Indexed: 11/16/2022]
Abstract
Numerous noncoding transcripts have been reported to correlate with cancer development and progression. Nevertheless, there remains a paucity of long noncoding RNAs (lncRNA) with well-elucidated functional roles. Here, we leverage the International Cancer Genome Consortium-Early Onset Prostate Cancer transcriptome and identify the previously uncharacterized lncRNA LINC00920 to be upregulated in prostate tumors. Phenotypic characterization of LINC00920 revealed its positive impact on cellular proliferation, colony formation, and migration. We demonstrate that LINC00920 transcription is directly activated by ERG, an oncogenic transcription factor overexpressed in 50% of prostate cancers. Chromatin isolation by RNA purification-mass spectrometry revealed the interaction of LINC00920 with the 14-3-3ε protein, leading to enhanced sequestration of tumor suppressive FOXO1. Altogether, our results provide a rationale on how ERG overexpression, partly by driving LINC00920 transcription, could confer survival advantage to prostate cancer cells and potentially prime PTEN-intact prostate cells for cellular transformation through FOXO inactivation. IMPLICATIONS: The study describes a novel lncRNA-mediated mechanism of regulating the FOXO signaling pathway and provides additional insight into the role of ERG in prostate cancer cells.
Collapse
Affiliation(s)
- Arlou Kristina Angeles
- Division of Cancer Genome Research, German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), and National Center for Tumor Diseases (NCT), Heidelberg, Germany.,Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Doreen Heckmann
- Division of Cancer Genome Research, German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), and National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - Niclas Flosdorf
- Division of Cancer Genome Research, German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), and National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - Stefan Duensing
- Department of Urology, Section of Molecular Urooncology, University Hospital Heidelberg, Heidelberg, Germany
| | - Holger Sültmann
- Division of Cancer Genome Research, German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), and National Center for Tumor Diseases (NCT), Heidelberg, Germany.
| |
Collapse
|
41
|
Isobe Y, Okumura M, McGregor LM, Brittain SM, Jones MD, Liang X, White R, Forrester W, McKenna JM, Tallarico JA, Schirle M, Maimone TJ, Nomura DK. Manumycin polyketides act as molecular glues between UBR7 and P53. Nat Chem Biol 2020; 16:1189-1198. [PMID: 32572277 PMCID: PMC7572527 DOI: 10.1038/s41589-020-0557-2] [Citation(s) in RCA: 93] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Accepted: 04/29/2020] [Indexed: 12/16/2022]
Abstract
Molecular glues are an intriguing therapeutic modality that harness small-molecules to induce interactions between proteins that typically do not interact. However, such molecules are rare and have been discovered fortuitously, thus limiting their potential as a general strategy for therapeutic intervention. We postulated that natural products bearing one or more electrophilic sites may be an unexplored source of new molecular glues, potentially acting through multi-covalent attachment. Using chemoproteomic platforms, we show that members of the manumycin family of polyketides, which bear multiple potentially reactive sites, target C374 of the putative E3 ligase UBR7 in breast cancer cells and engage in molecular glue interactions with the neo-substrate tumor-suppressor TP53, leading to p53 transcriptional activation and cell death. Our results reveal a novel anti-cancer mechanism of this natural product family and highlight the potential for combining chemoproteomics and multi-covalent natural products for the discovery of new molecular glues.
Collapse
Affiliation(s)
- Yosuke Isobe
- Department of Chemistry, University of California, Berkeley, Berkeley, CA, USA.,Novartis-Berkeley Center for Proteomics and Chemistry Technologies, Berkeley, CA, USA
| | - Mikiko Okumura
- Department of Chemistry, University of California, Berkeley, Berkeley, CA, USA.,Novartis-Berkeley Center for Proteomics and Chemistry Technologies, Berkeley, CA, USA
| | - Lynn M McGregor
- Novartis Institutes for BioMedical Research, Cambridge, MA, USA
| | | | - Michael D Jones
- Novartis Institutes for BioMedical Research, Cambridge, MA, USA
| | - Xiaoyou Liang
- Novartis Institutes for BioMedical Research, Cambridge, MA, USA
| | - Ross White
- Department of Chemistry, University of California, Berkeley, Berkeley, CA, USA.,Novartis-Berkeley Center for Proteomics and Chemistry Technologies, Berkeley, CA, USA
| | | | - Jeffrey M McKenna
- Novartis-Berkeley Center for Proteomics and Chemistry Technologies, Berkeley, CA, USA.,Novartis Institutes for BioMedical Research, Cambridge, MA, USA
| | - John A Tallarico
- Novartis-Berkeley Center for Proteomics and Chemistry Technologies, Berkeley, CA, USA.,Novartis Institutes for BioMedical Research, Cambridge, MA, USA
| | - Markus Schirle
- Novartis-Berkeley Center for Proteomics and Chemistry Technologies, Berkeley, CA, USA.,Novartis Institutes for BioMedical Research, Cambridge, MA, USA
| | - Thomas J Maimone
- Department of Chemistry, University of California, Berkeley, Berkeley, CA, USA. .,Novartis-Berkeley Center for Proteomics and Chemistry Technologies, Berkeley, CA, USA.
| | - Daniel K Nomura
- Department of Chemistry, University of California, Berkeley, Berkeley, CA, USA. .,Novartis-Berkeley Center for Proteomics and Chemistry Technologies, Berkeley, CA, USA. .,Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA. .,Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA, USA. .,Innovative Genomics Institute, Berkeley, CA, USA.
| |
Collapse
|
42
|
Vafaei H, Kavari G, Izadi HR, Zare Dorahi Z, Dianatpour M, Daneshparvar A, Jamhiri I. Wi-Fi (2.4 GHz) affects anti-oxidant capacity, DNA repair genes expression and, apoptosis in pregnant mouse placenta. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2020; 23:833-840. [PMID: 32695301 PMCID: PMC7351435 DOI: 10.22038/ijbms.2020.40184.9512] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Accepted: 12/25/2019] [Indexed: 01/09/2023]
Abstract
OBJECTIVES The placenta provides nutrients and oxygen to embryo and removes waste products from embryo's blood. As far as we know, the effects of exposure to Wi-Fi (2.4 GHz) signals on placenta have not been evaluated. Hence, we examined the effect of prenatal exposure to Wi-Fi signals on anti-oxidant capacity, expressions of CDKNA1, and GADD45a as well as apoptosis in placenta and pregnancy outcome. MATERIALS AND METHODS Pregnant mice were exposed to Wi-Fi signal (2.4 GHz) for 2 and 4 hr. Placenta tissues were examined to measure the MDA and SOD levels. To measure SOD, CDKNA1, GADD45a, Bax, and Bcl-2 expressions were compared by real-time PCR analysis. TUNEL assay was used to assess apoptosis in placenta tissues. The results were analyzed by one-way analysis of variance (ANOVA) using Prism version 6.0 software. RESULTS MDA and SOD levels had significantly increased in exposed Wi-Fi signal groups (P-value< 0.05). Also, quantitative PCR experiment showed that SOD mRNA expression significantly increased in Wi-Fi signal groups. The data showed that CDKN1A and GADD45a genes were increased in Wi-Fi groups (P-value<0.05). The quantitative PCR and the TUNEL assay showed that apoptosis increased in Wi-Fi groups (P-value<0.05). CONCLUSION Our results provide evidence that Wi-Fi signals increase lipid peroxidation, SOD activity (oxidative stres), apoptosis and CDKN1A and GADD45a overexpression in mice placenta tissue. However, further experimental studies are warranted to investigate other genes and aspects of pregnancy to determine the role of Wi-Fi radiation on fertility and pregnancy.
Collapse
Affiliation(s)
- Homeira Vafaei
- Maternal Fetal Medicine Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ghazal Kavari
- Maternal Fetal Medicine Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Hamid Reza Izadi
- Shiraz Neuroscience Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Zahra Zare Dorahi
- Stem Cell Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mehdi Dianatpour
- Stem Cell Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Human Genetics, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Afrooz Daneshparvar
- Stem Cell Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Iman Jamhiri
- Stem Cell Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
43
|
Strauch BM, Hubele W, Hartwig A. Impact of Endocytosis and Lysosomal Acidification on the Toxicity of Copper Oxide Nano- and Microsized Particles: Uptake and Gene Expression Related to Oxidative Stress and the DNA Damage Response. NANOMATERIALS 2020; 10:nano10040679. [PMID: 32260290 PMCID: PMC7221514 DOI: 10.3390/nano10040679] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 03/23/2020] [Accepted: 03/30/2020] [Indexed: 01/04/2023]
Abstract
The toxicity of the copper oxide nanoparticles (CuO NP) has been attributed to the so-called "Trojan horse"-type mechanism, relying on the particle uptake and extensive intracellular release of copper ions, due to acidic pH in the lysosomes. Nevertheless, a clear distinction between extra- and intracellular-mediated effects is still missing. Therefore, the impact of the endocytosis inhibitor hydroxy-dynasore (OH-dyn), as well as bafilomycin A1 (bafA1), inhibiting the vacuolar type H+-ATPase (V-ATPase), on the cellular toxicity of nano- and microsized CuO particles, was investigated in BEAS 2 B cells. Selected endpoints were cytotoxicity, copper uptake, glutathione (GSH) levels, and the transcriptional DNA damage and (oxidative) stress response using the high-throughput reverse transcription quantitative polymerase chain reaction (RT-qPCR). OH-dyn markedly reduced intracellular copper accumulation in the cases of CuO NP and CuO MP; the modulation of gene expression, induced by both particle types affecting especially HMOX1, HSPA1A, MT1X, SCL30A1, IL8 and GADD45A, were completely abolished. BafA1 lowered the intracellular copper concentration in case of CuO NP and strongly reduced transcriptional changes, while any CuO MP-mediated effects were not affected by bafA1. In conclusion, the toxicity of CuO NP depended almost exclusively upon dynamin-dependent endocytosis and the intracellular release of redox-active copper ions due to lysosomal acidification, while particle interactions with cellular membranes appeared to be not relevant.
Collapse
|
44
|
Plastics in Cyanobacterial Blooms-Genotoxic Effects of Binary Mixtures of Cylindrospermopsin and Bisphenols in HepG2 Cells. Toxins (Basel) 2020; 12:toxins12040219. [PMID: 32244372 PMCID: PMC7232240 DOI: 10.3390/toxins12040219] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 03/26/2020] [Accepted: 03/27/2020] [Indexed: 12/28/2022] Open
Abstract
Ever-expanding environmental pollution is causing a rise in cyanobacterial blooms and the accumulation of plastics in water bodies. Consequently, exposure to mixtures of cyanotoxins and plastic-related contaminants such as bisphenols (BPs) is of increasing concern. The present study describes genotoxic effects induced by co-exposure to one of the emerging cyanotoxins-cylindrospermopsin (CYN)-(0.5 µg/mL) and BPs (bisphenol A (BPA), S (BPS), and F (BPF); (10 µg/mL)) in HepG2 cells after 24 and 72 h of exposure. The cytotoxicity was evaluated with an MTS assay and genotoxicity was assessed through the measurement of the induction of DNA double strand breaks (DSB) with the γH2AX assay. The deregulation of selected genes (xenobiotic metabolic enzyme genes, DNA damage, and oxidative response genes) was assessed using qPCR. The results showed a moderate reduction of cell viability and induction of DSBs after 72 h of exposure to the CYN/BPs mixtures and CYN alone. None of the BPs alone reduced cell viability or induced DSBs. No significant difference was observed between CYN and CYN/BPs exposed cells, except with CYN/BPA, where the antagonistic activity of BPA against CYN was indicated. The deregulation of some of the tested genes (CYP1A1, CDKN1A, GADD45A, and GCLC) was more pronounced after exposure to the CYN/BPs mixtures compared to single compounds, suggesting additive or synergistic action. The present study confirms the importance of co-exposure studies, as our results show pollutant mixtures to induce effects different from those confirmed for single compounds.
Collapse
|
45
|
Interplay between BRCA1 and GADD45A and Its Potential for Nucleotide Excision Repair in Breast Cancer Pathogenesis. Int J Mol Sci 2020; 21:ijms21030870. [PMID: 32013256 PMCID: PMC7037490 DOI: 10.3390/ijms21030870] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 01/26/2020] [Accepted: 01/27/2020] [Indexed: 12/12/2022] Open
Abstract
A fraction of breast cancer cases are associated with mutations in the BRCA1 (BRCA1 DNA repair associated, breast cancer type 1 susceptibility protein) gene, whose mutated product may disrupt the repair of DNA double-strand breaks as BRCA1 is directly involved in the homologous recombination repair of such DNA damage. However, BRCA1 can stimulate nucleotide excision repair (NER), the most versatile system of DNA repair processing a broad spectrum of substrates and playing an important role in the maintenance of genome stability. NER removes carcinogenic adducts of diol-epoxy derivatives of benzo[α]pyrene that may play a role in breast cancer pathogenesis as their accumulation is observed in breast cancer patients. NER deficiency was postulated to be intrinsic in stage I of sporadic breast cancer. BRCA1 also interacts with GADD45A (growth arrest and DNA damage-inducible protein GADD45 alpha) that may target NER machinery to actively demethylate genome sites in order to change the expression of genes that may be important in breast cancer. Therefore, the interaction between BRCA1 and GADD45 may play a role in breast cancer pathogenesis through the stimulation of NER, increasing the genomic stability, removing carcinogenic adducts, and the local active demethylation of genes important for cancer transformation.
Collapse
|
46
|
Lee D, Hokinson D, Park S, Elvira R, Kusuma F, Lee JM, Yun M, Lee SG, Han J. ER Stress Induces Cell Cycle Arrest at the G2/M Phase Through eIF2α Phosphorylation and GADD45α. Int J Mol Sci 2019; 20:E6309. [PMID: 31847234 PMCID: PMC6940793 DOI: 10.3390/ijms20246309] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 12/06/2019] [Accepted: 12/11/2019] [Indexed: 01/04/2023] Open
Abstract
Endoplasmic reticulum (ER) stress is known to influence various cellular functions, including cell cycle progression. Although it is well known how ER stress inhibits cell cycle progression at the G1 phase, the molecular mechanism underlying how ER stress induces G2/M cell cycle arrest remains largely unknown. In this study, we found that ER stress and subsequent induction of the UPR led to cell cycle arrest at the G2/M phase by reducing the amount of cyclin B1. Pharmacological inhibition of the IRE1α or ATF6α signaling did not affect ER stress-induced cell cycle arrest at the G2/M phase. However, when the alpha subunit of eukaryotic translation initiation factor 2 (eIF2α) phosphorylation was genetically abrogated, the cell cycle progressed without arresting at the G2/M phase after ER stress. GEO database analysis showed that growth arrest and DNA-damage-inducible protein α (Gadd45α) were induced in an eIF2a phosphorylation-dependent manner, which was confirmed in this study. Knockdown of GADD45α abrogated cell cycle arrest at the G2/M phase upon ER stress. Finally, the cell death caused by ER stress significantly reduced when GADD45α expression was knocked down. In conclusion, GADD45α is a key mediator of ER stress-induced growth arrest via regulation of the G2/M transition and cell death through the eIF2α signaling pathway.
Collapse
Affiliation(s)
- Duckgue Lee
- Soonchunhyang Institute of Medi-bio Science (SIMS), Soonchunhyang University, Cheonan-si, Chungcheongnam-do 31151, Korea; (D.L.); (D.H.); (S.P.); (R.E.); (F.K.); (J.-M.L.)
| | - Daniel Hokinson
- Soonchunhyang Institute of Medi-bio Science (SIMS), Soonchunhyang University, Cheonan-si, Chungcheongnam-do 31151, Korea; (D.L.); (D.H.); (S.P.); (R.E.); (F.K.); (J.-M.L.)
| | - Soyoung Park
- Soonchunhyang Institute of Medi-bio Science (SIMS), Soonchunhyang University, Cheonan-si, Chungcheongnam-do 31151, Korea; (D.L.); (D.H.); (S.P.); (R.E.); (F.K.); (J.-M.L.)
| | - Rosalie Elvira
- Soonchunhyang Institute of Medi-bio Science (SIMS), Soonchunhyang University, Cheonan-si, Chungcheongnam-do 31151, Korea; (D.L.); (D.H.); (S.P.); (R.E.); (F.K.); (J.-M.L.)
| | - Fedho Kusuma
- Soonchunhyang Institute of Medi-bio Science (SIMS), Soonchunhyang University, Cheonan-si, Chungcheongnam-do 31151, Korea; (D.L.); (D.H.); (S.P.); (R.E.); (F.K.); (J.-M.L.)
| | - Ji-Min Lee
- Soonchunhyang Institute of Medi-bio Science (SIMS), Soonchunhyang University, Cheonan-si, Chungcheongnam-do 31151, Korea; (D.L.); (D.H.); (S.P.); (R.E.); (F.K.); (J.-M.L.)
| | - Miyong Yun
- Department of Bioindustry and Bioresource Engineering, College of Life Sciences, Sejong University, Seoul 02447, Korea;
| | - Seok-Geun Lee
- KHU-KIST Department of Converging Science & Technology, Department of Science in Korean Medicine, and Bionanocomposite Research Center, Kyung Hee Univerisity, 26 Kyungheedae-ro, Seoul 02447, Korea;
| | - Jaeseok Han
- Soonchunhyang Institute of Medi-bio Science (SIMS), Soonchunhyang University, Cheonan-si, Chungcheongnam-do 31151, Korea; (D.L.); (D.H.); (S.P.); (R.E.); (F.K.); (J.-M.L.)
| |
Collapse
|
47
|
Bang BR, Han KH, Seo GY, Croft M, Kang YJ. The protein tyrosine kinase SYK regulates the alternative p38 activation in liver during acute liver inflammation. Sci Rep 2019; 9:17838. [PMID: 31780731 PMCID: PMC6882802 DOI: 10.1038/s41598-019-54335-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Accepted: 11/11/2019] [Indexed: 12/02/2022] Open
Abstract
Two distinct p38 signaling pathways, classical and alternative, have been identified to regulate inflammatory responses in host defense and disease development. The role of alternative p38 activation in liver inflammation is elusive, while classical p38 signaling in hepatocytes plays a role in regulating the induction of cell death in autoimmune-mediated acute liver injury. In this study, we found that a mutation of alternative p38 in mice augmented the severity of acute liver inflammation. Moreover, TNF-induced hepatocyte death was augmented by a mutation of alternative p38, suggesting that alternative p38 signaling in hepatocytes contributed more significantly to the pathology of acute liver injury. Furthermore, SYK-Vav-1 signaling regulates alternative p38 activation and the downregulation of cell death in hepatocytes. Therefore, it is suggested that alternative p38 signaling in the liver plays a critical role in the induction and subsequent pathological changes of acute liver injury. Collectively, our results imply that p38 signaling in hepatocytes plays a crucial role to prevent excessive liver injury by regulating the induction of cell death and inflammation.
Collapse
Affiliation(s)
- Bo-Ram Bang
- Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, CA, 92037, USA
- Department of Medicine, Division of Gastrointestinal and Liver Diseases, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
| | - Kyung Ho Han
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Goo-Young Seo
- Division of Developmental Immunology, La Jolla Institute for Immunology, La Jolla, CA, 92037, USA
| | - Michael Croft
- Division of Immune Regulation, La Jolla Institute for Immunology, La Jolla, CA, 92037, USA
| | - Young Jun Kang
- Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, CA, 92037, USA.
- Molecular Medicine Research Institute, Sunnyvale, CA, 94085, USA.
| |
Collapse
|
48
|
Hercog K, Maisanaba S, Filipič M, Sollner-Dolenc M, Kač L, Žegura B. Genotoxic activity of bisphenol A and its analogues bisphenol S, bisphenol F and bisphenol AF and their mixtures in human hepatocellular carcinoma (HepG2) cells. THE SCIENCE OF THE TOTAL ENVIRONMENT 2019; 687:267-276. [PMID: 31207516 DOI: 10.1016/j.scitotenv.2019.05.486] [Citation(s) in RCA: 110] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 05/24/2019] [Accepted: 05/31/2019] [Indexed: 05/25/2023]
Abstract
The use of bisphenol A (BPA) in manufacturing of plastics is being gradually replaced by presumably safer analogues such as bisphenol S (BPS), bisphenol F (BPF) and bisphenol AF (BPAF). Despite their widespread occurrence in the environment, there is a knowledge gap in their toxicological profiles. We investigated cytotoxic/genotoxic effects as well as changes in the expression of selected genes involved in the xenobiotic metabolism, response to oxidative stress and DNA damage upon exposure to BPs and their mixtures in human hepatocellular carcinoma HepG2 cells. BPS and BPF slightly decreased the viability of HepG2 cells, while BPAF was the most cytotoxic compound tested. BPA, BPF and BPAF induced the formation of DNA double strand breaks determined with γH2AX assay, while BPS was inactive (5-20 μg/mL). All four BPs up-regulated the expression of CYP1A1 and UGT1A1, while BPS up-regulated and BPAF down-regulated also the expression of GST1A. Only BPA up-regulated oxidative stress responsive gene GCLC, while BPAF up-regulated the expression of CDKN1A and GADD45a. At concentrations relevant for human exposure (ng/mL range) BPA and its analogues as individual compounds and in mixtures did not exert genotoxic activity, whereas BPA and BPAF as well as the mixtures up-regulated the expressions of CYP1A1 and UGT1A1.
Collapse
Affiliation(s)
- Klara Hercog
- Department of Genetic Toxicology and Cancer Biology, National Institute of Biology, Ljubljana, Slovenia; Jozef Stefan International Postgraduate School, Ljubljana, Slovenia
| | - Sara Maisanaba
- Area of Toxicology, Department of Nutrition and Bromatology, Toxicology and Legal Medicine, Faculty of Pharmacy, University of Sevilla, Spain; Area of Toxicology, Department of Molecular Biology and Biochemistry Engineering, University Pablo de Olavide, Sevilla, Spain
| | - Metka Filipič
- Department of Genetic Toxicology and Cancer Biology, National Institute of Biology, Ljubljana, Slovenia
| | | | - Lidija Kač
- Department of Genetic Toxicology and Cancer Biology, National Institute of Biology, Ljubljana, Slovenia; Faculty of Pharmacy, University of Ljubljana, Ljubljana, Slovenia
| | - Bojana Žegura
- Department of Genetic Toxicology and Cancer Biology, National Institute of Biology, Ljubljana, Slovenia.
| |
Collapse
|
49
|
Li XL, Li GH, Fu J, Fu YW, Zhang L, Chen W, Arakaki C, Zhang JP, Wen W, Zhao M, Chen WV, Botimer GD, Baylink D, Aranda L, Choi H, Bechar R, Talbot P, Sun CK, Cheng T, Zhang XB. Highly efficient genome editing via CRISPR-Cas9 in human pluripotent stem cells is achieved by transient BCL-XL overexpression. Nucleic Acids Res 2019; 46:10195-10215. [PMID: 30239926 PMCID: PMC6212847 DOI: 10.1093/nar/gky804] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Accepted: 08/28/2018] [Indexed: 12/12/2022] Open
Abstract
Genome editing of human induced pluripotent stem cells (iPSCs) is instrumental for functional genomics, disease modeling, and regenerative medicine. However, low editing efficiency has hampered the applications of CRISPR–Cas9 technology in creating knockin (KI) or knockout (KO) iPSC lines, which is largely due to massive cell death after electroporation with editing plasmids. Here, we report that the transient delivery of BCL-XL increases iPSC survival by ∼10-fold after plasmid transfection, leading to a 20- to 100-fold increase in homology-directed repair (HDR) KI efficiency and a 5-fold increase in non-homologous end joining (NHEJ) KO efficiency. Treatment with a BCL inhibitor ABT-263 further improves HDR efficiency by 70% and KO efficiency by 40%. The increased genome editing efficiency is attributed to higher expressions of Cas9 and sgRNA in surviving cells after electroporation. HDR or NHEJ efficiency reaches 95% with dual editing followed by selection of cells with HDR insertion of a selective gene. Moreover, KO efficiency of 100% can be achieved in a bulk population of cells with biallelic HDR KO followed by double selection, abrogating the necessity for single cell cloning. Taken together, these simple yet highly efficient editing strategies provide useful tools for applications ranging from manipulating human iPSC genomes to creating gene-modified animal models.
Collapse
Affiliation(s)
- Xiao-Lan Li
- State Key Laboratory of Experimental Hematology, Tianjin 300020, China.,Institute of Hematology and Blood Disease Hospital, Tianjin 300020, China
| | - Guo-Hua Li
- State Key Laboratory of Experimental Hematology, Tianjin 300020, China.,Institute of Hematology and Blood Disease Hospital, Tianjin 300020, China
| | - Juan Fu
- Liaoning Provincial Key Laboratory of Cerebral Diseases, Institute for Brain Disorders, Dalian Medical University, Dalian 116044, China.,Department of Obstetrics and Gynecology, the First Affiliated Hospital of Dalian Medical University, Dalian 116044, China
| | - Ya-Wen Fu
- State Key Laboratory of Experimental Hematology, Tianjin 300020, China.,Institute of Hematology and Blood Disease Hospital, Tianjin 300020, China
| | - Lu Zhang
- State Key Laboratory of Experimental Hematology, Tianjin 300020, China.,Institute of Hematology and Blood Disease Hospital, Tianjin 300020, China
| | - Wanqiu Chen
- Department of Medicine, Loma Linda University, Loma Linda, CA 92350, USA
| | - Cameron Arakaki
- Department of Medicine, Loma Linda University, Loma Linda, CA 92350, USA
| | - Jian-Ping Zhang
- State Key Laboratory of Experimental Hematology, Tianjin 300020, China.,Institute of Hematology and Blood Disease Hospital, Tianjin 300020, China.,CAMS Key Laboratory of Gene Therapy for Blood Diseases, Tianjin 300020, China
| | - Wei Wen
- State Key Laboratory of Experimental Hematology, Tianjin 300020, China.,Institute of Hematology and Blood Disease Hospital, Tianjin 300020, China
| | - Mei Zhao
- State Key Laboratory of Experimental Hematology, Tianjin 300020, China.,Institute of Hematology and Blood Disease Hospital, Tianjin 300020, China
| | | | - Gary D Botimer
- Department of Orthopaedic Surgery, Loma Linda University, Loma Linda, CA 92350, USA
| | - David Baylink
- Department of Medicine, Loma Linda University, Loma Linda, CA 92350, USA
| | - Leslie Aranda
- Department of Medicine, Loma Linda University, Loma Linda, CA 92350, USA
| | - Hannah Choi
- Department of Medicine, Loma Linda University, Loma Linda, CA 92350, USA
| | - Rachel Bechar
- UCR Stem Cell Center and Core, University of California at Riverside, Riverside, CA 92521, USA
| | - Prue Talbot
- UCR Stem Cell Center and Core, University of California at Riverside, Riverside, CA 92521, USA
| | - Chang-Kai Sun
- Liaoning Provincial Key Laboratory of Cerebral Diseases, Institute for Brain Disorders, Dalian Medical University, Dalian 116044, China.,Research & Educational Center for the Control Engineering of Translational Precision Medicine (R-ECCE-TPM), School of Biomedical Engineering, Faculty of Electronic Information and Electrical Engineering, Dalian University of Technology, Dalian 116024, China.,State Key Laboratory of Fine Chemicals, Dalian R&D Center for Stem Cell and Tissue Engineering, Dalian University of Technology, Dalian 116024, China
| | - Tao Cheng
- State Key Laboratory of Experimental Hematology, Tianjin 300020, China.,Institute of Hematology and Blood Disease Hospital, Tianjin 300020, China.,Research & Educational Center for the Control Engineering of Translational Precision Medicine (R-ECCE-TPM), School of Biomedical Engineering, Faculty of Electronic Information and Electrical Engineering, Dalian University of Technology, Dalian 116024, China.,Center for Stem Cell Medicine, Chinese Academy of Medical Sciences, Tianjin 300020, China.,Department of Stem Cell & Regenerative Medicine, Peking Union Medical College, Tianjin 300020, China.,Collaborative Innovation Center for Cancer Medicine, Tianjin 300020, China
| | - Xiao-Bing Zhang
- State Key Laboratory of Experimental Hematology, Tianjin 300020, China.,Institute of Hematology and Blood Disease Hospital, Tianjin 300020, China.,Department of Medicine, Loma Linda University, Loma Linda, CA 92350, USA.,Center for Stem Cell Medicine, Chinese Academy of Medical Sciences, Tianjin 300020, China.,Department of Stem Cell & Regenerative Medicine, Peking Union Medical College, Tianjin 300020, China
| |
Collapse
|
50
|
Noh H, Shoemaker JE, Gunawan R. Network perturbation analysis of gene transcriptional profiles reveals protein targets and mechanism of action of drugs and influenza A viral infection. Nucleic Acids Res 2019; 46:e34. [PMID: 29325153 PMCID: PMC5887474 DOI: 10.1093/nar/gkx1314] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Accepted: 12/22/2017] [Indexed: 12/12/2022] Open
Abstract
Genome-wide transcriptional profiling provides a global view of cellular state and how this state changes under different treatments (e.g. drugs) or conditions (e.g. healthy and diseased). Here, we present ProTINA (Protein Target Inference by Network Analysis), a network perturbation analysis method for inferring protein targets of compounds from gene transcriptional profiles. ProTINA uses a dynamic model of the cell-type specific protein-gene transcriptional regulation to infer network perturbations from steady state and time-series differential gene expression profiles. A candidate protein target is scored based on the gene network's dysregulation, including enhancement and attenuation of transcriptional regulatory activity of the protein on its downstream genes, caused by drug treatments. For benchmark datasets from three drug treatment studies, ProTINA was able to provide highly accurate protein target predictions and to reveal the mechanism of action of compounds with high sensitivity and specificity. Further, an application of ProTINA to gene expression profiles of influenza A viral infection led to new insights of the early events in the infection.
Collapse
Affiliation(s)
- Heeju Noh
- Institute for Chemical and Bioengineering, ETH Zurich, Zurich 8093, Switzerland.,Swiss Institute of Bioinformatics, Lausanne 1015, Switzerland
| | - Jason E Shoemaker
- Department of Chemical and Petroleum Engineering, University of Pittsburgh, Pittsburgh, PA 15261, USA.,Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Rudiyanto Gunawan
- Institute for Chemical and Bioengineering, ETH Zurich, Zurich 8093, Switzerland.,Swiss Institute of Bioinformatics, Lausanne 1015, Switzerland
| |
Collapse
|