51
|
Tamtaji OR, Razavi ZS, Razzaghi N, Aschner M, Barati E, Mirzaei H. Quercetin and Glioma: Which signaling pathways are involved? Curr Mol Pharmacol 2022; 15:962-968. [DOI: 10.2174/1874467215666220211094136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 10/20/2021] [Accepted: 12/06/2021] [Indexed: 11/22/2022]
Abstract
Abstract:
Gliomas are the most common brain tumors. These tumors commonly exhibit continuous growth without invading surrounding brain tissues. Dominant remedial approaches suffer limited therapy and survival rates. Although some progress has been made in conventional glioma treatments, these breakthroughs have not yet proven sufficient for treating this malignancy. The remedial options are limited given gliomas' aggressive metastasis and drug resistance. Quercetin, a flavonoid, is an anti-oxidative, anti-allergic, antiviral, anti-inflammatory, and anticancer compound. Multiple lines of evidence have shown that Quercetin has anti-tumor effects, documenting this natural compound exerts its pharmacological effects by targeting a variety of cellular and molecular processes, i.e., apoptosis, metastasis, and autophagy. Herein, we summarize various cellular and molecular pathways that are affected by Quercetin in gliomas.
Collapse
Affiliation(s)
- Omid Reza Tamtaji
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, I.R. Iran
| | - Zahra Sadat Razavi
- Student Research Committee, Kashan University of Medical Sciences, Kashan, I.R. Iran
| | - Nazanin Razzaghi
- Laboratory Sciences Research Centre, School of Medicine, Golestan University of Medical Sciences, Gorgan, Iran
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, 10461, United States
| | - Erfaneh Barati
- Anatomical Sciences Research Center, Kashan University of Medical Sciences, Kashan, Iran
| | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, I.R. Iran
| |
Collapse
|
52
|
Montazeri-Najafabady N, Dabbaghmanesh MH, Nasimi N, Sohrabi Z, Estedlal A, Asmarian N. Importance of TP53 codon 72 and intron 3 duplication 16 bp polymorphisms and their haplotypes in susceptibility to sarcopenia in Iranian older adults. BMC Geriatr 2022; 22:103. [PMID: 35123410 PMCID: PMC8818191 DOI: 10.1186/s12877-022-02765-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 01/13/2022] [Indexed: 12/13/2022] Open
Abstract
Abstract
Background
Sarcopenia is described as age-related progressive skeletal muscle failure that results in marked reduction in the patient’s independence and life quality. In this study, we explored the association of TP53 exon 4 Arg72pro (rs1042522) and Intron 3 16-bp Del/Ins (rs17878362) polymorphisms and their haplotypes with sarcopenia, anthropometric, body composition and biochemical parameters.
Methods
A total of 254 older individuals (65 sarcopenic and 189 healthy) were recruited in this research and genotyped by PCR–RFLP. Linear regression was applied to find the correlation between TP53 polymorphism, and biochemical and anthropometric parameters. The correlation between TP53 polymorphism and haplotypes and the risk of sarcopenia was investigated by logistic regression.
Results
Arg/Pro genotype carriers was at a lower (ORadj = 0.175, 95% CI = 0.068 – 0.447; P < 0.001) risk of sarcopenia compared to the Arg/Arg group. In haplotypes analysis, Arg-Ins (ORadj: 0.484, 95% CI = 0.231 – 1.011, P = 0.043) and Pro-Ins (ORadj: 0.473, 95% CI = 0.210 – 1.068, P = 0.022) haplotypes showed decreased risk of developing sarcopenia. Moreover, in the case of codon 72 polymorphism, skeletal muscle mass, appendicular lean mass (ALM), skeletal muscle mass index (SMI), hand grip strength and Triglycerides, for Intron 3 16-bp Del/Ins polymorphism, albumin, calcium, cholesterol, and LDL were different, and for the haplotypes, skeletal muscle mass, SMI, ALM, HDL and triglycerides were significantly different between groups.
Conclusions
We suggested that the Arg/Pro genotype of the codon 72 polymorphism in exon 4 of TP53, and Arginine-Insertion and Proline-Insertion haplotypes might decrease the risk of sarcopenia in Iranian older adults.
Collapse
|
53
|
Abstract
Melanoma is a relentless type of skin cancer which involves myriad signaling pathways which regulate many cellular processes. This makes melanoma difficult to treat, especially when identified late. At present, therapeutics include chemotherapy, surgical resection, biochemotherapy, immunotherapy, photodynamic and targeted approaches. These interventions are usually administered as either a single-drug or in combination, based on tumor location, stage, and patients' overall health condition. However, treatment efficacy generally decreases as patients develop treatment resistance. Genetic profiling of melanocytes and the discovery of novel molecular factors involved in the pathogenesis of melanoma have helped to identify new therapeutic targets. In this literature review, we examine several newly approved therapies, and briefly describe several therapies being assessed for melanoma. The goal is to provide a comprehensive overview of recent developments and to consider future directions in the field of melanoma.
Collapse
Affiliation(s)
- Pavan Kumar Dhanyamraju
- Department of Pediatrics and Pharmacology, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
- Pavan Kumar Dhanyamraju, Department of Pediatrics and Pharmacology, Pennsylvania State University College of Medicine, Hershey, PA17033, USA. Tel: +1-6096474712, E-mail:
| | - Trupti N. Patel
- School of Biosciences and Technology, Vellore Institute of Technology, Vellore Campus, Vellore, Tamil Nadu 632014, India
| |
Collapse
|
54
|
Mei Y, Liang D, Wang T, Yu D. Gaining insights into relevance across cancers based on mutation features of TP53 gene. Biochem Biophys Rep 2021; 28:101165. [PMID: 34786492 PMCID: PMC8579115 DOI: 10.1016/j.bbrep.2021.101165] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Revised: 10/26/2021] [Accepted: 10/27/2021] [Indexed: 11/26/2022] Open
Abstract
The tumor suppressor gene TP53, one of the most frequently mutated genes, is recognized as the guardian of genome and can provide a significant barrier to neoplastic transformation and tumor progression. Traditional theory believes that TP53 mutations are equal among cancer types. However, to date, no study has explored the TP53 mutation profile from a holistic and systematic standpoint to discovery its relevance and feature with cancers. Mutation signature, an unbiased approach to identify the mutational processes, can be a potent indicator for exploring mutation-driven tumor occurrence and progression. In this research, several features such as hotspots, mutability and mutation signature of somatic TP53 mutations derived from 18 types of cancer tissues from cBioPortal were analyzed and manifested the organizational preference among cancers. Mutation signatures found in almost all cancer types were Signature 6 related to mismatch repair deficiency, and Signature 1 that reflects the natural decomposition of 5-methylcytosine into thymine associated with aging. Meanwhile, several signatures of TP53 mutations displayed tissue-selective. Mutations enriched in bladder, skin, lung cancer were associated with signatures of APOBEC activity (Signature 2 and 13), alkylating agents (Signature 11), and tobacco smoke (Signature 4), respectively. Moreover, Signature 4 and 29 associated with tobacco smoking or chewing found in lung, sarcoma, esophageal, and head and neck cancer may be related to their smoking history. In addition, several digestive cancers, including colorectal, stomach, pancreatic and esophageal cancers, showed the high correlation in context and mutation signature profiles. Our study suggests that the tissue-selective activity of mutational processes would reflect the tissue-specific enrichment of TP53 mutations and provides a new perspective to understand the relevance of diverse diseases based on the spectrum of TP53 mutations. TP53 mutations display the tissue-selective among cancers. Mutation signature of TP53 reflects the tissue-selective activity of mutational processes among cancers. Digestive cancers show the high correlation based on TP53 mutation signature.
Collapse
Affiliation(s)
- Yue Mei
- Center for Translational Medicine, Second Military Medical University, Shanghai, 200433, PR China
| | - Dong Liang
- Center for Translational Medicine, Second Military Medical University, Shanghai, 200433, PR China
| | - Tengjiao Wang
- Center for Translational Medicine, Second Military Medical University, Shanghai, 200433, PR China
| | - Dong Yu
- Center for Translational Medicine, Second Military Medical University, Shanghai, 200433, PR China
| |
Collapse
|
55
|
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
|
56
|
Genetic Predisposition to Persistent Human Papillomavirus-Infection and Virus-Induced Cancers. Microorganisms 2021; 9:microorganisms9102092. [PMID: 34683414 PMCID: PMC8539927 DOI: 10.3390/microorganisms9102092] [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: 09/16/2021] [Revised: 10/01/2021] [Accepted: 10/01/2021] [Indexed: 11/17/2022] Open
Abstract
Human papillomaviruses (HPVs) are the most common sexually transmitted pathogens worldwide and among the more than 200 identified HPV types, approximately 15 high risk (HR-HPV) types are oncogenic, being strongly associated with the development of cervical cancer, anogenital cancers and an increasing fraction of head and neck squamous cell carcinomas (HNSCC). HPV-associated cervix cancer accounts for 83% of HPV-attributable cancers, and more than two-thirds of those cases occur in developing countries. Despite the high frequency of HPV infections, in most cases, the virus is cleared by the host immune response and only a small proportion of infected individuals develop persistent infections that can result in malignant transformation, indicating that other elements, including biological, genetic and environmental factors may influence the individual susceptibility to HPV-associated cancers. Previous studies have quantified that heritability, in the form of genetic variants, common in the general population, is implicated in nearly 30% of cervical cancers and a large number of studies conducted across various populations have identified genetic variants that appear to be associated with genes that predispose or protect the host to HPV infections thereby affecting individual susceptibility to HPV-associated cancers. In this article, we provide an overview of gene association studies on HPV-associated cancers with emphasis on genome-wide association study (GWAS) that have identified novel genetic factors linked to HPV infection or HPV-associated cancers.
Collapse
|
57
|
Siemer S, Fauth T, Scholz P, Al-Zamel Y, Khamis A, Gül D, Freudelsperger L, Wollenberg B, Becker S, Stauber RH, Hagemann J. Profiling Cisplatin Resistance in Head and Neck Cancer: A Critical Role of the VRAC Ion Channel for Chemoresistance. Cancers (Basel) 2021; 13:4831. [PMID: 34638315 PMCID: PMC8508519 DOI: 10.3390/cancers13194831] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 09/23/2021] [Accepted: 09/24/2021] [Indexed: 12/11/2022] Open
Abstract
Treatment success of head and neck cancers (HNSCC) is often hindered by tumor relapses due to therapy resistances. This study aimed at profiling cisplatin resistance mechanisms and identifying biomarkers potentially suitable as drug targets and for patient stratification. Bioinformatic analyses of suggested resistance factors in a cohort of 565 HNSCC patients identified the VRAC ion channel as a clinically relevant indicator for recurrent diseases following radiochemotherapy (p = 0.042). Other drug import/export transporters, such as CTR1, OCT1, or MRP1, were found to be less relevant. To experimentally verify VRAC's critical role for cisplatin resistance, we used CRISPR/Cas9 knockout resulting in cisplatin-resistant HNSCC cells, which could be resensitized by VRAC expression. Next-generation sequencing further underlined VRAC's importance and identified VRAC-regulated signaling networks, potentially also contributing to cisplatin resistance. CTR1, OCT1, or MRP1 did not contribute to increased cisplatin resistance. In addition to two-dimensional HNSCC models, three-dimensional tumor spheroid cultures confirmed VRAC's unique role for cisplatin sensitivity. Here, resistance correlated with DNA damage and downstream apoptosis. The cisplatin specificity of the identified VRAC pathway was verified by testing paclitaxel and doxorubicin. Our results were independently confirmed in naturally occurring, cisplatin-resistant HNSCC cancer cell models. Collectively, we here demonstrate VRAC's role for cisplatin resistance in HNSCC and its relevance as a potential drug target and/or prognostic biomarker for chemotherapy resistance.
Collapse
Affiliation(s)
- Svenja Siemer
- Department of Otorhinolaryngology Head and Neck Surgery, Molecular and Cellular Oncology, University Medical Center, 55131 Mainz, Germany; (S.S.); (Y.A.-Z.); (A.K.); (D.G.); (L.F.); (S.B.); (J.H.)
| | - Torsten Fauth
- BRAIN Biotech AG, 64673 Zwingenberg, Germany; (T.F.); (P.S.)
| | - Paul Scholz
- BRAIN Biotech AG, 64673 Zwingenberg, Germany; (T.F.); (P.S.)
| | - Yara Al-Zamel
- Department of Otorhinolaryngology Head and Neck Surgery, Molecular and Cellular Oncology, University Medical Center, 55131 Mainz, Germany; (S.S.); (Y.A.-Z.); (A.K.); (D.G.); (L.F.); (S.B.); (J.H.)
| | - Aya Khamis
- Department of Otorhinolaryngology Head and Neck Surgery, Molecular and Cellular Oncology, University Medical Center, 55131 Mainz, Germany; (S.S.); (Y.A.-Z.); (A.K.); (D.G.); (L.F.); (S.B.); (J.H.)
| | - Désirée Gül
- Department of Otorhinolaryngology Head and Neck Surgery, Molecular and Cellular Oncology, University Medical Center, 55131 Mainz, Germany; (S.S.); (Y.A.-Z.); (A.K.); (D.G.); (L.F.); (S.B.); (J.H.)
| | - Laura Freudelsperger
- Department of Otorhinolaryngology Head and Neck Surgery, Molecular and Cellular Oncology, University Medical Center, 55131 Mainz, Germany; (S.S.); (Y.A.-Z.); (A.K.); (D.G.); (L.F.); (S.B.); (J.H.)
| | - Barbara Wollenberg
- Department of Otorhinolaryngology Head and Neck Surgery, University Hospital Klinikum Rechts der Isar, 81675 Munich, Germany;
| | - Sven Becker
- Department of Otorhinolaryngology Head and Neck Surgery, Molecular and Cellular Oncology, University Medical Center, 55131 Mainz, Germany; (S.S.); (Y.A.-Z.); (A.K.); (D.G.); (L.F.); (S.B.); (J.H.)
- Department of Otorhinolaryngology, University Medical Center Tuebingen, 72076 Tuebingen, Germany
| | - Roland H. Stauber
- Department of Otorhinolaryngology Head and Neck Surgery, Molecular and Cellular Oncology, University Medical Center, 55131 Mainz, Germany; (S.S.); (Y.A.-Z.); (A.K.); (D.G.); (L.F.); (S.B.); (J.H.)
| | - Jan Hagemann
- Department of Otorhinolaryngology Head and Neck Surgery, Molecular and Cellular Oncology, University Medical Center, 55131 Mainz, Germany; (S.S.); (Y.A.-Z.); (A.K.); (D.G.); (L.F.); (S.B.); (J.H.)
| |
Collapse
|
58
|
Doğan T, Atas H, Joshi V, Atakan A, Rifaioglu A, Nalbat E, Nightingale A, Saidi R, Volynkin V, Zellner H, Cetin-Atalay R, Martin M, Atalay V. CROssBAR: comprehensive resource of biomedical relations with knowledge graph representations. Nucleic Acids Res 2021; 49:e96. [PMID: 34181736 PMCID: PMC8450100 DOI: 10.1093/nar/gkab543] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Revised: 04/11/2021] [Accepted: 06/10/2021] [Indexed: 12/11/2022] Open
Abstract
Systemic analysis of available large-scale biological/biomedical data is critical for studying biological mechanisms, and developing novel and effective treatment approaches against diseases. However, different layers of the available data are produced using different technologies and scattered across individual computational resources without any explicit connections to each other, which hinders extensive and integrative multi-omics-based analysis. We aimed to address this issue by developing a new data integration/representation methodology and its application by constructing a biological data resource. CROssBAR is a comprehensive system that integrates large-scale biological/biomedical data from various resources and stores them in a NoSQL database. CROssBAR is enriched with the deep-learning-based prediction of relationships between numerous data entries, which is followed by the rigorous analysis of the enriched data to obtain biologically meaningful modules. These complex sets of entities and relationships are displayed to users via easy-to-interpret, interactive knowledge graphs within an open-access service. CROssBAR knowledge graphs incorporate relevant genes-proteins, molecular interactions, pathways, phenotypes, diseases, as well as known/predicted drugs and bioactive compounds, and they are constructed on-the-fly based on simple non-programmatic user queries. These intensely processed heterogeneous networks are expected to aid systems-level research, especially to infer biological mechanisms in relation to genes, proteins, their ligands, and diseases.
Collapse
Affiliation(s)
- Tunca Doğan
- Department of Computer Engineering, Hacettepe University, Ankara 06800, Turkey
- Institute of Informatics, Hacettepe University, Ankara 06800, Turkey
- Cancer Systems Biology Laboratory, Graduate School of Informatics, METU, Ankara 06800, Turkey
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL–EBI), Hinxton, Cambridgeshire CB10 1SD, UK
| | - Heval Atas
- Cancer Systems Biology Laboratory, Graduate School of Informatics, METU, Ankara 06800, Turkey
| | - Vishal Joshi
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL–EBI), Hinxton, Cambridgeshire CB10 1SD, UK
| | - Ahmet Atakan
- Department of Computer Engineering, METU, Ankara 06800, Turkey
- Department of Computer Engineering, EBYU, Erzincan 24002, Turkey
| | - Ahmet Sureyya Rifaioglu
- Department of Computer Engineering, METU, Ankara 06800, Turkey
- Department of Computer Engineering, İskenderun Technical University, Hatay 31200, Turkey
| | - Esra Nalbat
- Cancer Systems Biology Laboratory, Graduate School of Informatics, METU, Ankara 06800, Turkey
| | - Andrew Nightingale
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL–EBI), Hinxton, Cambridgeshire CB10 1SD, UK
| | - Rabie Saidi
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL–EBI), Hinxton, Cambridgeshire CB10 1SD, UK
| | - Vladimir Volynkin
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL–EBI), Hinxton, Cambridgeshire CB10 1SD, UK
| | - Hermann Zellner
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL–EBI), Hinxton, Cambridgeshire CB10 1SD, UK
| | - Rengul Cetin-Atalay
- Cancer Systems Biology Laboratory, Graduate School of Informatics, METU, Ankara 06800, Turkey
- Section of Pulmonary and Critical Care Medicine, University of Chicago, Chicago, IL 60637, USA
| | - Maria Martin
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL–EBI), Hinxton, Cambridgeshire CB10 1SD, UK
| | - Volkan Atalay
- Department of Computer Engineering, METU, Ankara 06800, Turkey
| |
Collapse
|
59
|
De Souza C, Madden J, Koestler DC, Minn D, Montoya DJ, Minn K, Raetz AG, Zhu Z, Xiao WW, Tahmassebi N, Reddy H, Nelson N, Karnezis AN, Chien J. Effect of the p53 P72R Polymorphism on Mutant TP53 Allele Selection in Human Cancer. J Natl Cancer Inst 2021; 113:1246-1257. [PMID: 33555293 PMCID: PMC8633460 DOI: 10.1093/jnci/djab019] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 01/06/2021] [Accepted: 02/03/2021] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND TP53 mutations occur in more than 50% of cancers. We sought to determine the effect of the intragenic P72R single nucleotide polymorphism (SNP; rs1042522) on the oncogenic properties of mutant p53. METHODS P72R allelic selection in tumors was determined from genotype calls and a Gaussian distributed mixture model. The SNP effect on mutant p53 was determined in p53-negative cancer cell lines. RNA-sequencing, chromatin immunoprecipitation, and survival analysis were performed to describe the SNP effect. All statistical tests were 2-sided. RESULTS Among 409 patients with germline heterozygous P72R SNP who harbored somatic mutations in TP53, we observed a selection bias against missense TP53 mutants encoding the P72 SNP (P = 1.64 x 10-13). Exogenously expressed hotspot p53 mutants with the P72 SNP were negatively selected in cancer cells. Gene expression analyses showed the enrichment of p53 pathway genes and inflammatory genes in cancer cells transduced with mutants encoding P72 SNP. Immune gene signature is enriched in patients harboring missense TP53 mutations with homozygous P72 SNP. These patients have improved overall survival as compared with those with the R72 SNP (P = .04). CONCLUSION This is the largest study demonstrating a selection against the P72 SNP. Missense p53 mutants with the P72 SNP retain partial wild-type tumor-suppressive functions, which may explain the selection bias against P72 SNP across cancer types. Ovarian cancer patients with the P72 SNP have a better prognosis than with the R72 SNP. Our study describes a previously unknown role through which the rs1042522 SNP modifies tumor suppressor activities of mutant p53 in patients.
Collapse
Affiliation(s)
- Cristabelle De Souza
- Department of Biochemistry and Molecular Medicine, UC Davis Medical Center, Sacramento, CA, USA
- University of New Mexico Biomedical Sciences Graduate Program, Albuquerque, NM, USA
- Current affiliation: Stanford University School of Medicine, Institute for Regenerative Medicine and Stem Cell Research, Stanford, CA, USA
| | - Jill Madden
- The Division of Genetics and Genomics, The Manton Center for Orphan Disease Research, Boston Children’s Hospital, Boston, MA, USA
| | - Devin C Koestler
- Department of Biostatistics and Data Science, Kansas University Medical Center, Kansas City, KS, USA
| | - Dennis Minn
- College of Information and Computer Sciences, University of Massachusetts, Amherst, MA, USA
| | - Dennis J Montoya
- Department of Biochemistry and Molecular Medicine, UC Davis Medical Center, Sacramento, CA, USA
| | - Kay Minn
- Novogene Corporation, Sacramento, CA, USA
| | - Alan G Raetz
- Department of Biochemistry and Molecular Medicine, UC Davis Medical Center, Sacramento, CA, USA
| | - Zheng Zhu
- Department of Biochemistry and Molecular Medicine, UC Davis Medical Center, Sacramento, CA, USA
| | - Wen-Wu Xiao
- Department of Biochemistry and Molecular Medicine, UC Davis Medical Center, Sacramento, CA, USA
| | - Neeki Tahmassebi
- Department of Biochemistry and Molecular Medicine, UC Davis Medical Center, Sacramento, CA, USA
| | - Harikumara Reddy
- Department of Biochemistry and Molecular Medicine, UC Davis Medical Center, Sacramento, CA, USA
| | - Nina Nelson
- Department of Biochemistry and Molecular Medicine, UC Davis Medical Center, Sacramento, CA, USA
| | - Anthony N Karnezis
- Department of Pathology and Laboratory Medicine, UC Davis Medical Center, Sacramento, CA, USA
| | - Jeremy Chien
- Department of Biochemistry and Molecular Medicine, UC Davis Medical Center, Sacramento, CA, USA
- Department of Obstetrics and Gynecology, UC Davis Medical Center, Sacramento, CA, USA
| |
Collapse
|
60
|
Hu G, Gao F, Wang G, Fang Y, Guo Y, Zhou J, Gu Y, Zhang C, Gao N, Wen Q, Qiao H. Use of proteomics to identify mechanisms of hepatocellular carcinoma with the CYP2D6*10 polymorphism and identification of ANGPTL6 as a new diagnostic and prognostic biomarker. J Transl Med 2021; 19:359. [PMID: 34412629 PMCID: PMC8375140 DOI: 10.1186/s12967-021-03038-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Accepted: 08/10/2021] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Although an association between the cytochrome P4502D6 (CYP2D6) *10 (100C>T) polymorphism and hepatocellular carcinoma (HCC) is known, the mechanism remains unclear. Here we aimed to explore mechanisms of CYP2D6*10 (100C>T) polymorphism conferring to HCC, and screen markers for HCC. METHODS Label-free global proteome profiling with 34 normal livers and peritumor tissue from 61 HCC patients was performed, and angiopoietin-like protein-6 (ANGPTL6) was evaluated in 2 liver samples validation cohorts and 2 blood specimens validation cohorts. RESULTS We found a significantly decreased frequency of TT in HCC patients which reduced HCC susceptibility by 69.2% and was accompanied by lowered enzymatic activity for CYP2D6. Proteomic analysis revealed 1342 differentially expressed proteins (DEPs) that were associated with HCC and 88 DEPs were identified as 100 TT-related proteins, likely underlying the susceptibility to HCC. Twenty-two upregulated DEPs and 66 downregulated DEPs were mainly related to lipid metabolism and the extracellular matrix, respectively. High ANGPTL6 was associated with a higher risk to HCC and worse prognosis. ANGPTL6 was both an independent risk factor and an independent prognostic factor for HCC and exhibited strong potential for predicting HCC occurrence, with comparable AUC values and higher sensitivity compared with alpha-fetoprotein. CONCLUSIONS The TT genotype-associated decreased risk of HCC appears to be related to lowered CYP2D6 activity and altered protein expression in the tumor microenvironment, and ANGPTL6 is a promising new diagnostic and prognostic biomarker for HCC. Our findings reveal new mechanistic insights for polymorphisms related to HCC risk and provide avenues for screening for HCC.
Collapse
Affiliation(s)
- Guiming Hu
- Institute of Clinical Pharmacology, Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Fei Gao
- Institute of Clinical Pharmacology, Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Guanzhe Wang
- Institute of Clinical Pharmacology, Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Yan Fang
- Institute of Clinical Pharmacology, Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Yuanyuan Guo
- Institute of Clinical Pharmacology, Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Jun Zhou
- Institute of Clinical Pharmacology, Zhengzhou University, Zhengzhou, 450052, Henan, China.,Affiliated People's Hospital of Zhengzhou University, Zhengzhou, China
| | - Yuhan Gu
- Institute of Clinical Pharmacology, Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Cunzhen Zhang
- Institute of Clinical Pharmacology, Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Na Gao
- Institute of Clinical Pharmacology, Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Qiang Wen
- Institute of Clinical Pharmacology, Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Hailing Qiao
- Institute of Clinical Pharmacology, Zhengzhou University, Zhengzhou, 450052, Henan, China.
| |
Collapse
|
61
|
Lodhi N, Singh R, Rajput SP, Saquib Q. SARS-CoV-2: Understanding the Transcriptional Regulation of ACE2 and TMPRSS2 and the Role of Single Nucleotide Polymorphism (SNP) at Codon 72 of p53 in the Innate Immune Response against Virus Infection. Int J Mol Sci 2021; 22:8660. [PMID: 34445373 PMCID: PMC8395432 DOI: 10.3390/ijms22168660] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Revised: 07/28/2021] [Accepted: 08/02/2021] [Indexed: 12/15/2022] Open
Abstract
Human ACE2 and the serine protease TMPRSS2 of novel SARS-CoV-2 are primary entry receptors in host cells. Expression of these genes at the transcriptional level has not been much discussed in detail. The ISRE elements of the ACE2 promoter are a binding site for the ISGF3 complex of the JAK/STAT signaling pathway. TMPRSS2, including IFNβ, STAT1, and STAT2, has the PARP1 binding site near to TSS either up or downstream promoter region. It is well documented that PARP1 regulates gene expression at the transcription level. Therefore, to curb virus infection, both promoting type I IFN signaling to boost innate immunity and prevention of virus entry by inhibiting PARP1, ACE2 or TMPRSS2 are safe options. Most importantly, our aim is to attract the attention of the global scientific community towards the codon 72 Single Nucleotide Polymorphism (SNP) of p53 and its underneath role in the innate immune response against SARS-CoV-2. Here, we discuss codon 72 SNP of human p53's role in the different innate immune response to restrict virus-mediated mortality rate only in specific parts of the world. In addition, we discuss potential targets and emerging therapies using bioengineered bacteriophage, anti-sense, or CRISPR strategies.
Collapse
Affiliation(s)
- Niraj Lodhi
- Clinical Research (Research and Development Division) miRNA Analytics LLC, Harlem Bio-Space, New York, NY 10027, USA
| | - Rubi Singh
- Department of Pharmacology, Weill Cornell Medicine, New York, NY 10065, USA;
| | | | - Quaiser Saquib
- Department of Zoology, College of Sciences, King Saud University, Riyadh 12372, Saudi Arabia;
| |
Collapse
|
62
|
Alsherbiny MA, Bhuyan DJ, Radwan I, Chang D, Li CG. Metabolomic Identification of Anticancer Metabolites of Australian Propolis and Proteomic Elucidation of Its Synergistic Mechanisms with Doxorubicin in the MCF7 Cells. Int J Mol Sci 2021; 22:ijms22157840. [PMID: 34360606 PMCID: PMC8346082 DOI: 10.3390/ijms22157840] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 07/18/2021] [Accepted: 07/19/2021] [Indexed: 12/11/2022] Open
Abstract
The combination of natural products with standard chemotherapeutic agents offers a promising strategy to enhance the efficacy or reduce the side effects of standard chemotherapy. Doxorubicin (DOX), a standard drug for breast cancer, has several disadvantages, including severe side effects and the development of drug resistance. Recently, we reported the potential bioactive markers of Australian propolis extract (AP-1) and their broad spectrum of pharmacological activities. In the present study, we explored the synergistic interactions between AP-1 and DOX in the MCF7 breast adenocarcinoma cells using different synergy quantitation models. Biochemometric and metabolomics-driven analysis was performed to identify the potential anticancer metabolites in AP-1. The molecular mechanisms of synergy were studied by analysing the apoptotic profile via flow cytometry, apoptotic proteome array and measuring the oxidative status of the MCF7 cells treated with the most synergistic combination. Furthermore, label-free quantification proteomics analysis was performed to decipher the underlying synergistic mechanisms. Five prenylated stilbenes were identified as the key metabolites in the most active AP-1 fraction. Strong synergy was observed when AP-1 was combined with DOX in the ratio of 100:0.29 (w/w) as validated by different synergy quantitation models implemented. AP-1 significantly enhanced the inhibitory effect of DOX against MCF7 cell proliferation in a dose-dependent manner with significant inhibition of the reactive oxygen species (p < 0.0001) compared to DOX alone. AP-1 enabled the reversal of DOX-mediated necrosis to programmed cell death, which may be advantageous to decline DOX-related side effects. AP-1 also significantly enhanced the apoptotic effect of DOX after 24 h of treatment with significant upregulation of catalase, HTRA2/Omi, FADD together with DR5 and DR4 TRAIL-mediated apoptosis (p < 0.05), contributing to the antiproliferative activity of AP-1. Significant upregulation of pro-apoptotic p27, PON2 and catalase with downregulated anti-apoptotic XIAP, HSP60 and HIF-1α, and increased antioxidant proteins (catalase and PON2) may be associated with the improved apoptosis and oxidative status of the synergistic combination-treated MCF7 cells compared to the mono treatments. Shotgun proteomics identified 21 significantly dysregulated proteins in the synergistic combination-treated cells versus the mono treatments. These proteins were involved in the TP53/ATM-regulated non-homologous end-joining pathway and double-strand breaks repairs, recruiting the overexpressed BRCA1 and suppressed RIF1 encoded proteins. The overexpression of UPF2 was noticed in the synergistic combination treatment, which could assist in overcoming doxorubicin resistance-associated long non-coding RNA and metastasis of the MCF7 cells. In conclusion, we identified the significant synergy and highlighted the key molecular pathways in the interaction between AP-1 and DOX in the MCF7 cells together with the AP-1 anticancer metabolites. Further in vivo and clinical studies are warranted on this synergistic combination.
Collapse
Affiliation(s)
- Muhammad A. Alsherbiny
- NICM Health Research Institute, Western Sydney University, Penrith, NSW 2751, Australia;
- Department of Pharmacognosy, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt
- Correspondence: (M.A.A.); (D.J.B.); (C.-G.L.)
| | - Deep J. Bhuyan
- NICM Health Research Institute, Western Sydney University, Penrith, NSW 2751, Australia;
- Correspondence: (M.A.A.); (D.J.B.); (C.-G.L.)
| | - Ibrahim Radwan
- Faculty of Science and Technology, University of Canberra, Canberra, ACT 2617, Australia;
| | - Dennis Chang
- NICM Health Research Institute, Western Sydney University, Penrith, NSW 2751, Australia;
| | - Chun-Guang Li
- NICM Health Research Institute, Western Sydney University, Penrith, NSW 2751, Australia;
- Correspondence: (M.A.A.); (D.J.B.); (C.-G.L.)
| |
Collapse
|
63
|
Wang Z, Liu Z, Mei J, Xu S, Liu Y. The next generation therapy for lung cancer: taking medicine by inhalation. NANOTECHNOLOGY 2021; 32:392002. [PMID: 34167099 DOI: 10.1088/1361-6528/ac0e68] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 06/23/2021] [Indexed: 06/13/2023]
Abstract
The inhalation administration method which has been applied to treat respiratory diseases has the characteristics of painlessness high efficiency and non-invasiveness, and the drug can also be targeted at the organ level first to reduce the loss of drug during circulation. Therefore, delivering medicine by inhalation administration has brought a new turnaround for lung cancer treatment. Herein from the perspective of combining traditional drug delivery design strategies with new drug delivery methods how to improve lung targeting efficiency and treatment efficacy is discussed. We also discuss the comparative advantages of inhaled drug delivery and traditional administration in the treatment of lung cancer such as intravenous injection. And the researches are divided into different forms of inhalation administration studied in the treatment of lung cancer in recent years, such as single-component loaded and multi-component loaded systems and their advantages. Finally, the obstacles of the application of carrier materials for inhalation administration and the prospects for improvement of lung cancer treatment methods are presented.
Collapse
Affiliation(s)
- Ziyao Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, People's Republic of China
- University of Chinese Academy of Sciences, Beijing 100049, People's Republic of China
| | - Zifan Liu
- Institute for Advanced Study, Shenzhen University, Shenzhen 518060, People's Republic of China
| | - Jie Mei
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, People's Republic of China
- University of Chinese Academy of Sciences, Beijing 100049, People's Republic of China
| | - Shanshan Xu
- Institute for Advanced Study, Shenzhen University, Shenzhen 518060, People's Republic of China
| | - Ying Liu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, People's Republic of China
- GBA National Institute for Nanotechnology Innovation, Guangdong 510700, People's Republic of China
| |
Collapse
|
64
|
Mancini F, Giorgini L, Teveroni E, Pontecorvi A, Moretti F. Role of Sex in the Therapeutic Targeting of p53 Circuitry. Front Oncol 2021; 11:698946. [PMID: 34307167 PMCID: PMC8298065 DOI: 10.3389/fonc.2021.698946] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Accepted: 06/16/2021] [Indexed: 12/03/2022] Open
Abstract
Sex profoundly affects cancer incidence and susceptibility to therapy, with sex hormones highly contributing to this disparity. Various studies and omics data suggest a relationship between sex and the oncosuppressor p53 circuitry, including its regulators MDM2 and MDM4. Association of this network with genetic variation underlies sex-related altered cancer risk, age of onset, and cancer sensitivity to therapy. Moreover, sex-related factors, mainly estrogenic hormones, can affect the levels and/or function of the p53 network both in hormone-dependent and independent cancer. Despite this evidence, preclinical and clinical studies aimed to evaluate p53 targeted therapy rarely consider sex and related factors. This review summarizes the studies reporting the relationship between sex and the p53 circuitry, including its associated regulators, MDM2 and MDM4, with particular emphasis on estrogenic hormones. Moreover, we reviewed the evaluation of sex/hormone in preclinical studies and clinical trials employing p53-target therapies, and discuss how patients’ sex and hormonal status could impact these therapeutic approaches.
Collapse
Affiliation(s)
- Francesca Mancini
- Research Unit on Human Reproduction, International Scientific Institute Paul VI, Fondazione Policlinico A. Gemelli, IRCCS, Rome, Italy
| | - Ludovica Giorgini
- Institute of Biochemistry and Cell Biology, National Research Council of Italy, Monterotondo, Italy.,Catholic University of the Sacred Heart of Rome, Fondazione Policlinico A. Gemelli, IRCCS, Rome, Italy
| | - Emanuela Teveroni
- Research Unit on Human Reproduction, International Scientific Institute Paul VI, Fondazione Policlinico A. Gemelli, IRCCS, Rome, Italy
| | - Alfredo Pontecorvi
- Catholic University of the Sacred Heart of Rome, Fondazione Policlinico A. Gemelli, IRCCS, Rome, Italy
| | - Fabiola Moretti
- Institute of Biochemistry and Cell Biology, National Research Council of Italy, Monterotondo, Italy
| |
Collapse
|
65
|
Zhao XM, Li YB, Sun P, Pu YD, Shan MJ, Zhang YM. Bioinformatics analysis of key biomarkers for retinoblastoma. J Int Med Res 2021; 49:3000605211022210. [PMID: 34187205 PMCID: PMC8371285 DOI: 10.1177/03000605211022210] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Objective To identify key genes involved in occurrence and development of retinoblastoma. Methods The microarray dataset, GSE5222, was downloaded from the gene expression omnibus (GEO) database. Differentially expressed genes (DEGs) between unilateral and bilateral retinoblastoma were identified and functional enrichment analysis performed. The protein–protein interaction (PPI) network was constructed and analysed by STRING and Cytoscape. Results DEGs were mainly associated with activation of cysteine-type endopeptidase activity involved in apoptotic process and small molecule catabolic process. Seven genes (WAS, GNB3, PTGER1, TACR1, GPR143, NPFF and CDKN2A) were identified as HUB genes. Conclusion Our research provides more understanding of the mechanisms of the disease at a molecular level and may help in the identification of novel biomarkers for retinoblastoma.
Collapse
Affiliation(s)
- Xin-Mei Zhao
- Ophthalmic Laboratory, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong, China
| | - Yuan-Bin Li
- Department of Ophthalmology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong, China
| | - Peng Sun
- Department of Ophthalmology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong, China
| | - Ya-di Pu
- Qingdao University, Qingdao, Shandong, China
| | - Meng-Jie Shan
- Department of Plastic Surgery, Peking Union Medical College Hospital, Beijing, China.,Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Yuan-Meng Zhang
- Department of Cardiology, The Third Medical Centre of Chinese PLA General Hospital, No. 69, Yongding Road, Hai Dian, Beijing, China
| |
Collapse
|
66
|
Silva SN, Gomes BC, André S, Félix A, Rodrigues AS, Rueff J. Male and female breast cancer: the two faces of the same genetic susceptibility coin. Breast Cancer Res Treat 2021; 188:295-305. [PMID: 33942220 PMCID: PMC8233260 DOI: 10.1007/s10549-021-06159-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Accepted: 02/20/2021] [Indexed: 02/02/2023]
Abstract
BACKGROUND Breast cancer (BC) is the most common cancer in women. In contrast, male BC is about 100 times less common than in women, being considered a rare disease. Male BC may be a distinctive subtype of BC and available data seems to indicate that male BC has a higher dependence on genetic variants than female BC. Nevertheless, the same prognostic and predictive markers are used to determine optimal management strategies for both male and female BC. Several studies have assessed the role of genetic polymorphisms (SNPs) in DNA repair genes in female BC susceptibility. However, data on male BC is scarce. Thus, the current study aimed to assess the role of SNPs in XRCC1, MUTYH and TP53 genes in a male cohort of BC, and, in addition, compare the male data with matched results previously genotyped in female BC patients. METHODS The male BC cohort was genotyped through Real-Time PCR using TaqMan Assays for several SNPs previously analysed in Portuguese female BC patients. RESULTS The results obtained indicate significant differences in BC susceptibility between males and females for the XRCC1 rs1799782, MUTYH rs3219489 and TP53 rs1042522 and rs8064946 variants. CONCLUSIONS In males, XRCC1 and TP53 variants, when in heterozygosity, seem to be related with lower susceptibility for BC, contrasting with higher susceptibility for a MUTYH variant in females. These findings may help to explain the difference in incidence of BC between the two sexes.
Collapse
Affiliation(s)
- Susana Nunes Silva
- Center for Toxicogenomics and Human Health, NOVA Medical School, NMS, Universidade Nova de Lisboa, Rua Câmara Pestana, No.6, 1169-056, Lisbon, Portugal.
| | - Bruno Costa Gomes
- Center for Toxicogenomics and Human Health, NOVA Medical School, NMS, Universidade Nova de Lisboa, Rua Câmara Pestana, No.6, 1169-056, Lisbon, Portugal
| | - Saudade André
- Department of Pathology, Portuguese Institute of Oncology of Lisbon, 1099-023, Lisbon, Portugal
| | - Ana Félix
- Department of Pathology, Portuguese Institute of Oncology of Lisbon, 1099-023, Lisbon, Portugal
- NOVA Medical School, Universidade Nova de Lisboa, 1169-056, Lisbon, Portugal
| | - António Sebastião Rodrigues
- Center for Toxicogenomics and Human Health, NOVA Medical School, NMS, Universidade Nova de Lisboa, Rua Câmara Pestana, No.6, 1169-056, Lisbon, Portugal
| | - José Rueff
- Center for Toxicogenomics and Human Health, NOVA Medical School, NMS, Universidade Nova de Lisboa, Rua Câmara Pestana, No.6, 1169-056, Lisbon, Portugal.
| |
Collapse
|
67
|
Gansmo LB, Lie BA, Mæhlen MT, Vatten L, Romundstad P, Hveem K, Lønning PE, Knappskog S. Polymorphisms in the TP53-MDM2-MDM4-axis in patients with rheumatoid arthritis. Gene 2021; 793:145747. [PMID: 34077778 DOI: 10.1016/j.gene.2021.145747] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 04/30/2021] [Accepted: 05/27/2021] [Indexed: 10/21/2022]
Abstract
BACKGROUND In addition to being a tumour suppressor, TP53 is a suppressor of inflammation, and dysfunction of this gene has been related to autoimmune diseases. Patients with autoimmunity, such as rheumatoid arthritis (RA) have an increased risk of certain cancers, like lymphomas, indicating that some underlying mechanisms may modulate risk of both cancers and autoimmunity. METHODS We genotyped 5 common genetic variants in TP53 and its main regulators MDM2 and MDM4 in a sample of 942 RA patients and 3,747 healthy controls, and mined previously published GWAS-data, to assess the potential impact of these variants on risk of RA. RESULTS For the TP53 Arg72Pro polymorphism (rs1042522), MDM4 SNP34091 (rs4245739) and MDM2 SNP285C (rs117039649), we found no association to risk of RA. For MDM2 SNP309 (rs2279744), the minor G-allele was associated with a reduced risk of RA (OR: 0.87; CI: 0.79-0.97). This association was also seen in genotype models (OR: 0.86; CI: 0.74-0.99 and OR: 0.79; CI 0.63-0.99; dominant and recessive model, respectively), but was not validated in a large GWAS data set. For MDM2 del1518 (rs3730485), the minor del-allele was associated with an increased risk of RA in the dominant model (OR: 1.18; CI: 1.02-1.38). Stratifying RA cases and controls into phylogenetic subgroups according to the combined genotypes of all three MDM2 polymorphism, we found individuals with the del158-285-309 genotype del/ins-G/G-T/T to have an increased risk of RA as compared to those with the ins/ins-G/G-G/G genotype (OR: 1.56; CI: 1.18-2.06) indicating opposite effects of the del1518 del-allele and the SNP309 G-allele. CONCLUSION We find a potential association between the MDM2 del1518 variant and RA, and indications that combinatorial genotypes and haplotypes in the MDM2 locus may be related to RA.
Collapse
Affiliation(s)
- Liv B Gansmo
- K.G. Jebsen Center for Genome-Directed Cancer Therapy, Department of Clinical Science, University of Bergen, Bergen, Norway; Department of Oncology, Haukeland University Hospital, Bergen, Norway
| | - Benedicte A Lie
- Department of Medical Genetics, Oslo University Hospital, Oslo, Norway
| | - Marthe T Mæhlen
- Department of Medical Genetics, Oslo University Hospital, Oslo, Norway; Department of Rheumatology, Diakonhjemmet Hospital, Oslo, Norway
| | - Lars Vatten
- Department of Public Health, Faculty of Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Pål Romundstad
- Department of Public Health, Faculty of Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Kristian Hveem
- K.G. Jebsen Center for Genetic Epidemiology, Department of Public Health, Faculty of Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Per E Lønning
- K.G. Jebsen Center for Genome-Directed Cancer Therapy, Department of Clinical Science, University of Bergen, Bergen, Norway; Department of Oncology, Haukeland University Hospital, Bergen, Norway
| | - Stian Knappskog
- K.G. Jebsen Center for Genome-Directed Cancer Therapy, Department of Clinical Science, University of Bergen, Bergen, Norway; Department of Oncology, Haukeland University Hospital, Bergen, Norway.
| |
Collapse
|
68
|
Rodriguez KD, Schneider KW, Suttman A, Garrington T, Jellins T, Tholen K, Francom CR, Herrmann BW. Pediatric Head and Neck Tumors Associated with Li-Fraumeni Syndrome. Ann Otol Rhinol Laryngol 2021:34894211014786. [PMID: 33971750 DOI: 10.1177/00034894211014786] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
INTRODUCTION Cancer predisposition syndromes are germline pathogenic variants in genes that greatly raise the risk of developing neoplastic diseases. One of the most well-known is Li-Fraumeni syndrome (LFS), which is due to pathogenic variants in the TP53 gene. Children with LFS have higher risks for multiple malignancies before adulthood, often with rare and aggressive subtypes. OBJECTIVE To examine head and neck manifestations of LFS in children treated at a tertiary children's hospital over a 20-year period. METHODS A retrospective review of LFS children with neoplastic disease presenting in traditional Otolaryngologic head and neck subsites from 2000 to 2019, with patient charts reviewed for relevant clinical, imaging, and operative data. RESULTS Of the 40 LFS patients initially identified, 27 neoplastic tumors were identified in 20 children within this cohort (20 primary, 7 second primary). Head and neck subsites aside from the brain or orbit were involved in 22% (6/27) of these tumors, representing 20% (4/20) of primary tumors and 29% (2/7) of second primary tumors. Both second primaries within the head and neck were within the radiation fields of the first primary tumor. The mean ages at primary and second primary diagnosis were 4.6 years (SD 3.5) and 12 years (SD 1.4), respectively. The male/female ratio was 1:6 among all patients with head and neck tumors. All 6 head and neck tumors were sarcomas. Rhabdomyosarcoma (N = 3, 50%) was the most common pathology, and the other 3 demonstrated rare tumor pathological subtypes (synovial cell sarcoma, pleomorphic myxoid liposarcoma, mandibular osteosarcoma). The neck was the most common subsite (75%) within this group for primary tumor presentation. CONCLUSION This study identifies a high potential for head and neck involvement in children with LFS, which has not been previously described in the literature. Otolaryngological care should be included in a multidisciplinary care team surveilling these patients.
Collapse
Affiliation(s)
- Kenny D Rodriguez
- Department of Otolaryngology-Head and Neck Surgery, University of Colorado School of Medicine and Children's Hospital Colorado, Aurora, CO, USA
| | - Kami Wolfe Schneider
- Department of Hematology, Oncology, Bone Marrow Transplantation, University of Colorado School of Medicine and Children's Hospital Colorado, Aurora, CO, USA
| | - Alexandra Suttman
- Department of Hematology, Oncology, Bone Marrow Transplantation, University of Colorado School of Medicine and Children's Hospital Colorado, Aurora, CO, USA
| | - Timothy Garrington
- Department of Hematology, Oncology, Bone Marrow Transplantation, University of Colorado School of Medicine and Children's Hospital Colorado, Aurora, CO, USA
| | | | - Kaitlyn Tholen
- Department of Otolaryngology-Head and Neck Surgery, University of Colorado School of Medicine and Children's Hospital Colorado, Aurora, CO, USA
| | - Christian R Francom
- Department of Otolaryngology-Head and Neck Surgery, University of Colorado School of Medicine and Children's Hospital Colorado, Aurora, CO, USA
| | - Brian W Herrmann
- Department of Otolaryngology-Head and Neck Surgery, University of Colorado School of Medicine and Children's Hospital Colorado, Aurora, CO, USA
| |
Collapse
|
69
|
Yi X, Zhou Q, Sui G, Ren G, Tan L, Li J, Lin J, Bao S. Interactions among variants in P53 apoptotic pathway genes are associated with neurologic deterioration and functional outcome after acute ischemic stroke. Brain Behav 2021; 11:e01492. [PMID: 31909567 PMCID: PMC8119796 DOI: 10.1002/brb3.1492] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 11/04/2019] [Accepted: 11/11/2019] [Indexed: 12/17/2022] Open
Abstract
OBJECTIVE Neurologic deterioration (ND) and functional outcome after ischemic stroke (IS) are not accurately predicted by clinical pictures on admission. The aim of present study was to investigate the association of variants in P53 apoptotic pathway genes with ND and functional outcome after IS. METHODS Genotypes of nine variants in apoptosis-relevant genes were measured in patients with acute IS. Gene-gene interactions were analyzed by generalized multifactor dimensionality reduction (GMDR). The primary outcome was ND. ND was diagnosed in patients who worsened ≥2 points (National Institutes of Health Stroke Scale [NIHSS] score) within the first 10 days of stroke onset. The secondary outcome was functional status at 90 days after IS as measured by modified Rankin Scale (mRS) score. RESULTS A total of 705 enrolled patients, ND occurred in 174 (24.7%) patients, and 184 (26.1%) patients were poor functional outcome (mRS score > 2). Although the nine variants were not significantly associated with ND and functional outcome by univariate analysis, there was a gene-gene interaction among P53rs1042522, MDM-2rs2279744, and MMP-9 rs3918242 using GMDR analysis. The high-risk interaction among the three variants was independently associated with higher risk of ND (HR, 2.04, 95% CI: 1.22-5.64, p = .018) and poor functional outcome (OR, 2.68, 95% CI: 1.68-7.86, p = .004) after adjusting for the covariates. CONCLUSION The interactions among P53 rs1042522, MDM-2 rs2279744, and MMP-9 rs3918242 may increase the risk of ND and poor functional outcome and may be considered as a genetic marker of predicting ND and poor functional outcome after stroke.
Collapse
Affiliation(s)
- Xingyang Yi
- Department of Neurology, The People's Hospital of Deyang City, Deyang, Sichuan, China
| | - Qiang Zhou
- Department of Neurology, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Guo Sui
- Nursing Department, People's Hospital of Deyang City, Deyang, Sichuan, China
| | - Gaoping Ren
- Department of Neurology, The People's Hospital of Deyang City, Deyang, Sichuan, China
| | - Lili Tan
- Nursing Department, People's Hospital of Deyang City, Deyang, Sichuan, China
| | - Jie Li
- Department of Neurology, The People's Hospital of Deyang City, Deyang, Sichuan, China
| | - Jing Lin
- Department of Neurology, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Shaozhi Bao
- Department of Neurology, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| |
Collapse
|
70
|
Zarei-Ghobadi M, Sheikhi M, Teymoori-Rad M, Yaslianifard S, Norouzi M, Yaslianifard S, Faraji R, Farahmand M, Bayat S, Jafari M, Mozhgani SH. HTLV-1-associated myelopathy/tropical spastic paraparesis (HAM/TSP) versus adult T-cell leukemia/lymphoma (ATLL). BMC Res Notes 2021; 14:109. [PMID: 33757561 PMCID: PMC7989087 DOI: 10.1186/s13104-021-05521-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 03/11/2021] [Indexed: 11/17/2022] Open
Abstract
Objectives Human T cell leukemia virus-1 (HTLV-1) infection may lead to one or both diseases including HTLV-1-associated myelopathy/tropical spastic paraparesis (HAM/TSP) or adult T cell leukemia lymphoma (ATLL). The complete interactions of the virus with host cells in both diseases is yet to be determined. This study aims to construct an interaction network for distinct signaling pathways in these diseases based on finding differentially expressed genes (DEGs) between HAM/TSP and ATLL. Results We identified 57 hub genes with higher criteria scores in the primary protein–protein interaction network (PPIN). The ontology-based enrichment analysis revealed following important terms: positive regulation of transcription from RNA polymerase II promoter, positive regulation of transcription from RNA polymerase II promoter involved in meiotic cell cycle and positive regulation of transcription from RNA polymerase II promoter by histone modification. The upregulated genes TNF, PIK3R1, HGF, NFKBIA, CTNNB1, ESR1, SMAD2, PPARG and downregulated genes VEGFA, TLR2, STAT3, TLR4, TP53, CHUK, SERPINE1, CREB1 and BRCA1 were commonly observed in all the three enriched terms in HAM/TSP vs. ATLL. The constructed interaction network was then visualized inside a mirrored map of signaling pathways for ATLL and HAM/TSP, so that the functions of hub genes were specified in both diseases. Supplementary Information The online version contains supplementary material available at 10.1186/s13104-021-05521-y.
Collapse
Affiliation(s)
| | - Mohsen Sheikhi
- Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran
| | - Majid Teymoori-Rad
- Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran
| | - Sahar Yaslianifard
- Department of Biochemistry, Faculty of Life Sciences of Islamic, Azad University, Tehran north branch, Tehran, Iran
| | - Mehdi Norouzi
- Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran.,Research Center for Clinical Virology, Tehran University of Medical Sciences, Tehran, Iran
| | - Somayeh Yaslianifard
- Department of Microbiology, School of Medicine, Alborz University of Medical Sciences, Karaj, Iran.,Dietary Supplements and Probiotic Research Center, Alborz University of Medical Sciences, Karaj, Iran
| | - Reza Faraji
- Department of Microbiology, School of Medicine, Alborz University of Medical Sciences, Karaj, Iran
| | - Mohammad Farahmand
- Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran
| | - Shiva Bayat
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohieddin Jafari
- Research Program in Systems Oncology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Sayed-Hamidreza Mozhgani
- Department of Microbiology, School of Medicine, Alborz University of Medical Sciences, Karaj, Iran. .,Non-Communicable Diseases Research Center, Alborz University of Medical Sciences, Karaj, Iran.
| |
Collapse
|
71
|
Goizueta AA, Estrada-Y-Martin RM, Cherian SV. Lung Cancer in Women: a Review. CURRENT PULMONOLOGY REPORTS 2021. [DOI: 10.1007/s13665-021-00270-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
72
|
Pouladi N, Shavali M, Abdolahi S. Combined Genotype Effects of TP53 and PAI-1 Polymorphisms in Breast Cancer Susceptibility: Multifactor Dimensionality Reduction and in silico Analysis. Hum Hered 2021; 85:51-60. [PMID: 33735891 DOI: 10.1159/000514398] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 01/12/2021] [Indexed: 11/19/2022] Open
Abstract
INTRODUCTION Breast cancer is a heterogeneous and multifactorial disease. TP53 and PAI-1 as important tumor suppressor genes are involved in the development, invasion, and metastasis of many cancers. This study's objective was to demonstrate the combined genotype effects of these 2 genes by investigating their single nucleotide polymorphisms. METHODS In this case-control study, 200 individuals with breast cancer and 179 healthy individuals were studied. The genotypes were determined using the tetra-ARMS method. For data analysis, MDR, online javstat statistics package, and SPSS v.24 software were used. Also, in silico studies on the estimated effects of each of these polymorphisms were performed. RESULTS We showed a novel gene-gene interaction of these 2 genes and demonstrated a strong synergistic interaction for TP53/PAI-1, moderate synergistic interaction for PAI-1/age, and correlation for TP53/age. On the other hand, there was no association between the allelic and genotype frequency alone and in combination, with case-control status, using the parametric method, between TP53 and PAI-1. DISCUSSION/CONCLUSION Our findings suggest that the polymorphism of codon 72 of the TP53 gene was significantly associated with tumor stage (p < 0.023). In conclusion, we showed a gene-gene interaction between TP53 and PAI-1, in combination, using the MDR method.
Collapse
Affiliation(s)
- Nasser Pouladi
- Department of Biology, Faculty of Science, Azarbaijan Shahid Madani University, Tabriz, Iran,
| | - Mojtaba Shavali
- Department of Biology, Faculty of Science, Azarbaijan Shahid Madani University, Tabriz, Iran
| | - Sepehr Abdolahi
- Department of Biology, Faculty of Science, Azarbaijan Shahid Madani University, Tabriz, Iran
| |
Collapse
|
73
|
Almeida A, Sánchez-Morán I, Rodríguez C. Mitochondrial-nuclear p53 trafficking controls neuronal susceptibility in stroke. IUBMB Life 2021; 73:582-591. [PMID: 33615665 PMCID: PMC8248069 DOI: 10.1002/iub.2453] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2020] [Revised: 12/30/2020] [Accepted: 12/31/2020] [Indexed: 12/12/2022]
Abstract
Stroke is a major cause of death and long-term disability in the adult. Neuronal apoptosis plays an essential role in the pathophysiology of ischemic brain damage and impaired functional recovery after stroke. The tumor suppressor protein p53 regulates key cellular processes, including cell cycle arrest, DNA repair, senescence, and apoptosis. Under cellular stress conditions, p53 undergoes post-translational modifications, which control protein localization, stability, and proapoptotic activity. After stroke, p53 rapidly accumulates in the ischemic brain, where it activates neuronal apoptosis through both transcriptional-dependent and -independent programs. Over the last years, subcellular localization of p53 has emerged as an important regulator of ischemia-induced neuronal apoptosis. Upon an ischemic insult, p53 rapidly translocates to the mitochondria and interacts with B-cell lymphoma-2 family proteins, which activate the mitochondrial apoptotic program, with higher efficacy than through its activity as a transcription factor. Moreover, the identification of a human single nucleotide polymorphism at codon 72 of the Tp53 gene that controls p53 mitochondrial localization and cell susceptibility to apoptosis supports the important role of the p53 mitochondrial program in neuronal survival and functional recovery after stroke. In this article, we review the relevance of mitochondrial and nuclear localization of p53 on neuronal susceptibility to cerebral ischemia and its impact on functional outcome of stroke patients.
Collapse
Affiliation(s)
- Angeles Almeida
- Institute of Functional Biology and Genomics, CSIC, University of Salamanca, Salamanca, Spain.,Institute of Biomedical Research of Salamanca, University Hospital of Salamanca, University of Salamanca, CSIC, Salamanca, Spain
| | - Irene Sánchez-Morán
- Institute of Functional Biology and Genomics, CSIC, University of Salamanca, Salamanca, Spain.,Institute of Biomedical Research of Salamanca, University Hospital of Salamanca, University of Salamanca, CSIC, Salamanca, Spain
| | - Cristina Rodríguez
- Institute of Functional Biology and Genomics, CSIC, University of Salamanca, Salamanca, Spain.,Institute of Biomedical Research of Salamanca, University Hospital of Salamanca, University of Salamanca, CSIC, Salamanca, Spain
| |
Collapse
|
74
|
Mihalcea CE, Moroşanu AM, Murăraşu D, Puiu L, Cinca SA, Voinea SC, Mirancea N. Analysis of TP53 gene and particular infrastructural alterations in invasive ductal mammary carcinoma. ROMANIAN JOURNAL OF MORPHOLOGY AND EMBRYOLOGY 2021; 61:441-447. [PMID: 33544795 PMCID: PMC7864295 DOI: 10.47162/rjme.61.2.13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
This study was conducted in order to determine the mutational status of TP53 gene and to determine some particular aspects from ultrastructural level in invasive mammary ductal carcinoma. The cellular signaling pathway involving the TP53 gene acts in biological deoxyribonucleic acid (DNA) repair processes and cell cycle arrest following a signal transmitted to the p53 protein when posttranslational changes occur in the cell due to stress induced in the cell by both intrinsic and extrinsic factors. Cellular stress activates the transcription factor function of the protein that initiates, as the case may be, either DNA repair or programmed cell death (apoptosis). The TP53 gene is commonly mutated in many human cancers and also has a highly polymorphic grade. To determine the mutational status of the exons 4–9 of the TP53 gene, we used extracted DNA from fresh breast tissue, and we analyzed it through direct sequencing. In mammary carcinoma, the mutation frequency of TP53 is running between 20–40% and, in regards the polymorphism, at least 14 different forms were identified, that are associated with cancer risk. The mutation type distribution showed a predominance of deletions and a reduced frequency of substitutions comparing with International Agency for Research on Cancer (IARC) database. Taken in consideration the importance of the tumor associated stroma in tumor development, we have also investigated some particular aspects at the infrastructural level of invasive mammary ductal carcinoma, notably concerning telocytes as tumor stroma interstitial cells by transmission electron microscopy analysis.
Collapse
Affiliation(s)
- Corina Elena Mihalcea
- Department of Plant and Animal Cytobiology, Institute of Biology Bucharest of Romanian Academy, Bucharest, Romania;
| | | | | | | | | | | | | |
Collapse
|
75
|
Ye Z, Xie T, Yan F, Wang L, Fang J, Wang Z, Hu F, Wang F, Fu Z. MiR-34a reverses radiation resistance on ECA-109 cells by inhibiting PI3K/AKT/mTOR signal pathway through downregulating the expression of SIRT1. Int J Radiat Biol 2021; 97:452-463. [PMID: 33507132 DOI: 10.1080/09553002.2021.1866225] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 12/10/2020] [Accepted: 12/14/2020] [Indexed: 12/15/2022]
Abstract
BACKGROUND Radiotherapy is an effective treatment for esophageal squamous cell carcinoma (ESCC). However, many ESCC patients relapsed after receiving radiotherapy due to the inherent resistance. The function of miR-34a and SIRT1, as well as the correlation between miR-34a and SIRT1 has been widely claimed in multiple types of malignant tumors. This study aimed to investigate the effects of miR-34a on radiation resistance against ESCC and the underlying mechanism. METHODS In this study, CCK8, flow cytometry, wounding healing assays, and cell clone formation assay were used to determine the in vitro anti-tumor effects of radiation on radiation-resistant ESCC cell line (rECA-109). The luciferase activity and Western Blot assays were used to investigate the relationship among miR-34a, SIRT1, and the anti-radiation resistant effects. The xenograft experiments were used to verify the important function of miR-34a and SIRT1 in radiation resistance against ESCC. The apoptosis state of tumor tissues was evaluated by TUNEL assay. RESULTS The introduction of miR-34a significantly induced the cell death and apoptosis of rECA-109 and inhibit the migration of rECA-109 treated by radiation. The anti-tumor effect was accompanied by the downregulation of SIRT1 and the inhibition of PI3K/AKT/mTOR signal pathway. The radiation resistance on rECA-109 cells was reversed by silencing SIRT1, accompanied by the PI3K/AKT/mTOR signal pathway inhibited. In vivo experiments revealed that the radiation resistance on ESCC was reversed by the introduction of miR-34a, the effect of which was promoted by the activation of SIRT1. CONCLUSION Our results showed that miR-34a could reverse the radiation resistance on rECA-109 cells by downregulating the expression of SIRT1through inhibiting the PI3K-AKT-mTOR signal pathway.
Collapse
Affiliation(s)
- Zhimin Ye
- Department of Radiation Oncology, Hangzhou City, China
- Key Laboratory of Head & Neck Cancer Translational Research of Zhejiang Pronvince, Hangzhou City, China
| | - Tieming Xie
- Department of Radiation Oncology, Hangzhou City, China
- Key Laboratory of Head & Neck Cancer Translational Research of Zhejiang Pronvince, Hangzhou City, China
| | - Fengqin Yan
- Department of Radiation Oncology, Hangzhou City, China
- Key Laboratory of Head & Neck Cancer Translational Research of Zhejiang Pronvince, Hangzhou City, China
| | - Lei Wang
- Department of Radiation Oncology, Hangzhou City, China
- Key Laboratory of Head & Neck Cancer Translational Research of Zhejiang Pronvince, Hangzhou City, China
| | - Jun Fang
- Department of Radiation Oncology, Hangzhou City, China
| | - Zhun Wang
- Department of Radiation Oncology, Hangzhou City, China
| | - Fujun Hu
- Department of Radiation Oncology, Hangzhou City, China
- Key Laboratory of Head & Neck Cancer Translational Research of Zhejiang Pronvince, Hangzhou City, China
| | - Fangzheng Wang
- Department of Radiation Oncology, Hangzhou City, China
- Key Laboratory of Head & Neck Cancer Translational Research of Zhejiang Pronvince, Hangzhou City, China
| | - Zhenfu Fu
- Department of Radiation Oncology, Hangzhou City, China
- Key Laboratory of Head & Neck Cancer Translational Research of Zhejiang Pronvince, Hangzhou City, China
| |
Collapse
|
76
|
Ma W, Zhao P, Zang L, Zhang K, Liao H, Hu Z. CircTP53 promotes the proliferation of thyroid cancer via targeting miR-1233-3p/MDM2 axis. J Endocrinol Invest 2021; 44:353-362. [PMID: 32500444 DOI: 10.1007/s40618-020-01317-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Accepted: 05/28/2020] [Indexed: 12/14/2022]
Abstract
BACKGROUND Follicular cells give rise to thyroid cancer. Worldwide thyroid cancer incidence continues to rise in recent decades but the mortality rate remains at a stable level. The discovery of novel molecular mechanisms in the pathogenesis of thyroid cancer will promote new diagnostic or therapeutic strategies. Circular RNA (circRNA) is a type of noncoding RNA which is characterized by the covalently closed loop and non-protein coding capacity. The abnormal expression of circRNAs is an important part during the pathogenesis and development of thyroid cancer. CircTP53 is a novel circRNA, and we aimed to investigate its function in the pathogenesis of thyroid cancer and to further demonstrate the underlying molecular mechanism. METHODS The levels of circTP53, miR-1233-3p, and other relative mRNA were analyzed by qRT-PCR. Protein levels were shown by Western blot. RNA-pulldown assay and luciferase assay were employed to examine the interaction between circTP53 and miR-1233-3p. Cell proliferation was analyzed by the MTT assay. RESULTS CircTP53 was a circRNA highly expressed in thyroid cancer tissues. CircTP53 promoted cell proliferation and cell viability of TPC-1 cells. Knockdown of circTP53 inhibited the expression of Mouse double minute 2 (MDM2) and increased the protein level of p53. CircTP53 acted as a target of miR-1233-3p to increase MDM2 expression. p53 expression in thyroid cancer tissue exhibited a negative correlation with circTP53 expression. CONCLUSION In thyroid cancer, overexpressed circTP53 decreased the protein level of p53 via targeting miR-1233-3p/MDM2 axis and promoted cancer cell proliferation.
Collapse
Affiliation(s)
- W Ma
- The Second Hospital of Hebei Medical University, No. 215 Heping West Road, Shijiazhuang, 050000, Hebei, China
| | - P Zhao
- The Second Hospital of Hebei Medical University, No. 215 Heping West Road, Shijiazhuang, 050000, Hebei, China
| | - L Zang
- The Second Hospital of Hebei Medical University, No. 215 Heping West Road, Shijiazhuang, 050000, Hebei, China
| | - K Zhang
- The Second Hospital of Hebei Medical University, No. 215 Heping West Road, Shijiazhuang, 050000, Hebei, China
| | - H Liao
- The Second Hospital of Hebei Medical University, No. 215 Heping West Road, Shijiazhuang, 050000, Hebei, China
| | - Z Hu
- The Second Hospital of Hebei Medical University, No. 215 Heping West Road, Shijiazhuang, 050000, Hebei, China.
| |
Collapse
|
77
|
Chang KF, Chang JT, Huang XF, Huang YC, Li CY, Weng JC, Hsiao CY, Hsu HJ, Tsai NM. Cedrus atlantica Extract Suppress Glioblastoma Growth through Promotion of Genotoxicity and Apoptosis: In Vitro and In Vivo Studies. Int J Med Sci 2021; 18:2417-2430. [PMID: 33967620 PMCID: PMC8100640 DOI: 10.7150/ijms.54468] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 04/07/2021] [Indexed: 01/31/2023] Open
Abstract
Glioblastoma (GBM) is the most common malignant primary brain tumor in humans, exhibiting highly infiltrative growth and drug resistance to conventional chemotherapy. Cedrus atlantica (CAt) extract has been shown to decrease postoperative pain and inhibit the growth of K562 leukemia cells. The aim of this study was to assess the anti-GBM activity and molecular mechanism of CAt extract in vitro and in vivo. The results showed that CAt extract greatly suppressed GBM cells both in vitro and in vivo and enhanced the survival rate in subcutaneous and orthotopic animal models. Moreover, CAt extract increased the level of ROS and induced DNA damage, resulting in cell cycle arrest at the G0/G1 phase and cell apoptosis. Western blotting results indicated that CAt extract regulates p53/p21 and CDK4/cyclin D1 protein expression and activates extrinsic and intrinsic apoptosis. Furthermore, CAt extract enhanced the cytotoxicity of Temozolomide and decreased AKT/mTOR signaling by combination treatment. In toxicity assays, CAt extract exhibited low cytotoxicity toward normal cells or organs in vitro and in vivo. CAt extract suppresses the growth of GBM by induction of genotoxicity and activation of apoptosis. The results of this study suggest that CAt extract can be developed as a therapeutic agent or adjuvant for GBM treatment in the future.
Collapse
Affiliation(s)
- Kai-Fu Chang
- Institute of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan, ROC.,Department of Medical Laboratory and Biotechnology, Chung Shan Medical University, Taichung 40201, Taiwan, ROC
| | - Jinghua Tsai Chang
- Institute of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan, ROC
| | - Xiao-Fan Huang
- Institute of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan, ROC.,Department of Medical Laboratory and Biotechnology, Chung Shan Medical University, Taichung 40201, Taiwan, ROC
| | - Ya-Chih Huang
- Institute of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan, ROC.,Department of Medical Laboratory and Biotechnology, Chung Shan Medical University, Taichung 40201, Taiwan, ROC
| | - Chia-Yu Li
- Department of Life and Death, Nanhua University, Chiayi 62249, Taiwan, ROC
| | - Jun-Cheng Weng
- Department of Medical Imaging and Radiological Sciences, Chang Gung University, Taoyuan 33302, Taiwan, ROC
| | - Chih-Yen Hsiao
- Division of Nephrology, Department of Internal Medicine, Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chia-Yi, 60002, Taiwan, ROC.,Department of Hospital and Health Care Administration, Chia Nan University of Pharmacy and Science, Tainan, 71710, Taiwan, ROC
| | - Hui-Ju Hsu
- Department of Medical Laboratory and Biotechnology, Chung Shan Medical University, Taichung 40201, Taiwan, ROC.,Clinical Laboratory, Chung Shan Medical University Hospital, Taichung 40201, Taiwan, ROC
| | - Nu-Man Tsai
- Department of Medical Laboratory and Biotechnology, Chung Shan Medical University, Taichung 40201, Taiwan, ROC.,Clinical Laboratory, Chung Shan Medical University Hospital, Taichung 40201, Taiwan, ROC
| |
Collapse
|
78
|
Liu Z, Xiao Z, Li M, Xiao Y, Wang X, He J, Li Y. Association Between Arg72Pro Polymorphism in TP53 and Malignant Abdominal Solid Tumor Risk in Hunan Children. Cancer Control 2021; 28:10732748211004880. [PMID: 33759598 PMCID: PMC8204553 DOI: 10.1177/10732748211004880] [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: 01/09/2021] [Revised: 01/09/2021] [Accepted: 02/26/2021] [Indexed: 02/05/2023] Open
Abstract
Pediatric solid tumors are heterogeneous and comprise various histological subtypes. TP53, a tumor suppressor, orchestrates the transcriptional activation of anti-cancer genes. The gene coding for this protein is highly polymorphic, and its mutations are associated with cancer development. The Arg72Pro polymorphism in TP53 has been associated with susceptibility to various types of cancer. Here, in this hospital-based study, we evaluated the association of this polymorphism with susceptibility toward malignant abdominal solid tumors in children in the Hunan province of China. We enrolled 162 patients with neuroblastoma, 60 patients with Wilms' tumor, and 28 patients with hepatoblastoma as well as 270 controls. Genotypes were determined using a TaqMan assay, and the strength of the association was assessed using an odds ratio, within a 95% confidence interval identified using logistic regression models. Our results showed that the Arg72Pro polymorphism did not exhibit significant association with susceptibility toward pediatric malignant abdominal solid tumors. Stratification analysis revealed that this polymorphism exerts weak sex- and age-specific effects on Wilms' tumor and hepatoblastoma susceptibility, respectively. Overall, our results indicate that the Arg72Pro polymorphism may have a marginal effect on susceptibility toward pediatric malignant abdominal solid tumors in Hunan, and this finding warrants further confirmation.
Collapse
Affiliation(s)
- Zan Liu
- Department of Pediatric Surgery, Hunan Children’s Hospital, Changsha, Hunan, China
| | - Zhenghui Xiao
- Emergency Center of Hunan Children’s Hospital, Changsha, Hunan, China
| | - Ming Li
- Department of Pediatric Surgery, Hunan Children’s Hospital, Changsha, Hunan, China
| | - Yaling Xiao
- Department of Pediatric Surgery, Hunan Children’s Hospital, Changsha, Hunan, China
| | - Xiyang Wang
- Department of Spine Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jing He
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Yong Li
- Department of Pediatric Surgery, Hunan Children’s Hospital, Changsha, Hunan, China
| |
Collapse
|
79
|
Lang GT, Shi JX, Huang L, Cao AY, Zhang CH, Song CG, Zhuang ZG, Hu X, Huang W, Shao ZM. Multiple cancer susceptible genes sequencing in BRCA-negative breast cancer with high hereditary risk. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:1417. [PMID: 33313162 PMCID: PMC7723566 DOI: 10.21037/atm-20-2999] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Background Hereditary factors contributed to breast cancer susceptibility. Low BRCA mutation prevalence was demonstrated in previous BRCA mutation screening in Chinese breast cancer patients. Multiple-gene sequencing may assist in discovering detrimental germline mutation in BRCA negative breast cancers. Methods A total of 384 Chinese subjects with any two of high-risk factors were recruited and screened by next-generation sequencing (NGS) for 30 cancer susceptible genes. Variants with a truncating, initiation codon or splice donor/acceptor effect, or with pathogenicity demonstrated in published literature were classified into pathogenic/likely-pathogenic mutations. Results In total, we acquired 39 (10.2%) patients with pathogenic/likely-pathogenic germline mutations, including one carrying two distinct mutations. Major mutant non-BRCA genes were MUTYH (n=11, 2.9%), PTCH1 (n=7, 1.8%), RET (n=6, 1.6%) and PALB2 (n=5, 1.3%). Other mutant genes included TP53 (n=3, 0.8%), RAD51D (n=2, 0.5%), CHEK2 (n=1, 0.3%), BRIP1 (n=1, 0.3%), CDH1 (n=1, 0.3%), MRE11 (n=1, 0.3%), RAD50 (n=1, 0.3%) and PALLD (n=1, 0.3%). A splicing germline mutation, MUTYH c.934-2A>G, was a hotspot (9/384, 2.3%) in Chinese breast cancer. Conclusions Among BRCA-negative breast cancer patients with high hereditary risk in China, 10.2% carried mutations in cancer associated susceptibility genes. MUTYH and PTCH1 had relatively high mutation rates (2.9% and 1.8%). Multigene testing contributes to understand genetic background of BRCA-negative breast cancer patients with high hereditary risk.
Collapse
Affiliation(s)
- Guan-Tian Lang
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jin-Xiu Shi
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, Chinese National Human Genome Center at Shanghai (CHGC) and Shanghai Industrial Technology Institute (SITI), Shanghai, China
| | - Liang Huang
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - A-Yong Cao
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Chen-Hui Zhang
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, Chinese National Human Genome Center at Shanghai (CHGC) and Shanghai Industrial Technology Institute (SITI), Shanghai, China
| | - Chuan-Gui Song
- Department of Breast Surgery, Affiliated Union Hospital, Fujian Medical University, Fuzhou, China
| | - Zhi-Gang Zhuang
- Department of Breast Surgery, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xin Hu
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Wei Huang
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, Chinese National Human Genome Center at Shanghai (CHGC) and Shanghai Industrial Technology Institute (SITI), Shanghai, China
| | - Zhi-Ming Shao
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
80
|
The P72R Polymorphism in R248Q/W p53 Mutants Modifies the Mutant Effect on Epithelial to Mesenchymal Transition Phenotype and Cell Invasion via CXCL1 Expression. Int J Mol Sci 2020; 21:ijms21218025. [PMID: 33126568 PMCID: PMC7662892 DOI: 10.3390/ijms21218025] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Revised: 10/21/2020] [Accepted: 10/21/2020] [Indexed: 12/18/2022] Open
Abstract
High-grade serous carcinoma (HGSC), the most lethal subtype of epithelial ovarian cancer (EOC), is characterized by widespread TP53 mutations (>90%), most of which are missense mutations (>70%). The objective of this study was to investigate differential transcriptional targets affected by a common germline P72R SNP (rs1042522) in two p53 hotspot mutants, R248Q and R248W, and identify the mechanism through which the P72R SNP affects the neomorphic properties of these mutants. Using isogenic cell line models, transcriptomic analysis, xenografts, and patient data, we found that the P72R SNP modifies the effect of p53 hotspot mutants on cellular morphology and invasion properties. Most importantly, RNA sequencing studies identified CXCL1 a critical factor that is differentially affected by P72R SNP in R248Q and R248W mutants and is responsible for differences in cellular morphology and functional properties observed in these p53 mutants. We show that the mutants with the P72 SNP promote a reversion of the EMT phenotype to epithelial characteristics, whereas its R72 counterpart promotes a mesenchymal transition via the chemokine CXCL1. These studies reveal a new role of the P72R SNP in modulating the neomorphic properties of p53 mutants via CXCL1, which has significant implications for tumor invasion and metastasis.
Collapse
|
81
|
Gebert M, Jaśkiewicz M, Moszyńska A, Collawn JF, Bartoszewski R. The Effects of Single Nucleotide Polymorphisms in Cancer RNAi Therapies. Cancers (Basel) 2020; 12:E3119. [PMID: 33113880 PMCID: PMC7694039 DOI: 10.3390/cancers12113119] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 10/14/2020] [Accepted: 10/23/2020] [Indexed: 12/12/2022] Open
Abstract
Tremendous progress in RNAi delivery methods and design has allowed for the effective development of siRNA-based therapeutics that are currently under clinical investigation for various cancer treatments. This approach has the potential to revolutionize cancer therapy by providing the ability to specifically downregulate or upregulate the mRNA of any protein of interest. This exquisite specificity, unfortunately, also has a downside. Genetic variations in the human population are common because of the presence of single nucleotide polymorphisms (SNPs). SNPs lead to synonymous and non-synonymous changes and they occur once in every 300 base pairs in both coding and non-coding regions in the human genome. Much less common are the somatic mosaicism variations associated with genetically distinct populations of cells within an individual that is derived from postzygotic mutations. These heterogeneities in the population can affect the RNAi's efficacy or more problematically, which can lead to unpredictable and sometimes adverse side effects. From a more positive viewpoint, both SNPs and somatic mosaicisms have also been implicated in human diseases, including cancer, and these specific changes could offer the ability to effectively and, more importantly, selectively target the cancer cells. In this review, we discuss how SNPs in the human population can influence the development and success of novel anticancer RNAi therapies and the importance of why SNPs should be carefully considered.
Collapse
Affiliation(s)
- Magdalena Gebert
- Department of Biology and Pharmaceutical Botany, Medical University of Gdańsk, 80-416 Gdańsk, Poland; (M.G.); (M.J.); (A.M.)
| | - Maciej Jaśkiewicz
- Department of Biology and Pharmaceutical Botany, Medical University of Gdańsk, 80-416 Gdańsk, Poland; (M.G.); (M.J.); (A.M.)
| | - Adrianna Moszyńska
- Department of Biology and Pharmaceutical Botany, Medical University of Gdańsk, 80-416 Gdańsk, Poland; (M.G.); (M.J.); (A.M.)
| | - James F. Collawn
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35294, USA;
| | - Rafał Bartoszewski
- Department of Biology and Pharmaceutical Botany, Medical University of Gdańsk, 80-416 Gdańsk, Poland; (M.G.); (M.J.); (A.M.)
| |
Collapse
|
82
|
Sun C, Li M, Feng Y, Sun F, Zhang L, Xu Y, Lu S, Zhu J, Huang J, Wang J, Hu Y, Zhang Y. MDM2-P53 Signaling Pathway-Mediated Upregulation of CDC20 Promotes Progression of Human Diffuse Large B-Cell Lymphoma. Onco Targets Ther 2020; 13:10475-10487. [PMID: 33116627 PMCID: PMC7575066 DOI: 10.2147/ott.s253758] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Accepted: 07/14/2020] [Indexed: 01/03/2023] Open
Abstract
Background Cell-division cycle 20 (CDC20) is overexpressed in a variety of tumor cells and is negatively regulated by wild-type p53 (wtp53). Our previous study uncovered that CDC20 was upregulated and associated with poor outcome in diffuse large B-cell lymphoma (DLBCL) based on bioinformatics analysis. Dysregulation of the MDM2-p53 is a major mechanism to promote DLBCL. Thus, we hypothesized that CDC20 could be a downstream gene of the MDM2-p53 signaling pathway. However, the clinical significance and mechanistic role of a novel MDM2-p53-CDC20 signaling pathway in DLBCL have still remained unclear. Materials and Methods RT-qPCR was performed in MDM2 knocked down (KD) and control (Ctrl) OCI-Ly3/OCI-Ly10 cells to investigate whether CDC20 was a downstream gene of the MDM2-p53 pathway. The effects of CDC20 on cell proliferation, cell cycle and apoptosis were assessed, as well as the role of CDC20 in suppressing tumorigenicity in vivo. Furthermore, we also investigated the roles of CDC20 and MDM2 in progression of DLBCL and the underlying mechanisms. Results The results of RT-qPCR revealed that CDC20 was downregulated while TP53 was upregulated in MDM2 KD OCI-Ly3 and OCI-Ly10 cells. It was unveiled that the expression levels of CDC20 and MDM2 were upregulated in DLBCL tissues and cells, and high CDC20 expression was correlated with adverse clinical features and poor outcome. Functional assays showed that downregulation of CDC20 could inhibit proliferation, induce apoptosis and cell cycle arrest in vitro. In addition, inactivation of the MDM2-p53 pathway by downregulation of MDM2 restored wtp53 expression level and reduced CDC20 protein level in OCI-Ly3 and OCI-Ly10 cells. Besides, targeting CDC20 was found to suppress tumorigenesis of DLBCL in vivo. Conclusion CDC20 was identified as a key downstream gene of the MDM2-p53 signaling pathway in DLBCL and may be used as a novel target gene to guide therapeutic applications.
Collapse
Affiliation(s)
- Chengtao Sun
- Department of Hematology, Tianjin Medical University Cancer Institute and Hospital; National Clinical Research Center for Cancer; Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin, People's Republic of China
| | - Mengzhen Li
- Department of Hematology, Tianjin Medical University Cancer Institute and Hospital; National Clinical Research Center for Cancer; Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin, People's Republic of China
| | - Yanfen Feng
- Sun Yat-Sen University Cancer Center; State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong Province, People's Republic of China.,Department of Pathology, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong Province, People's Republic of China
| | - Feifei Sun
- Sun Yat-Sen University Cancer Center; State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong Province, People's Republic of China.,Department of Pediatric Oncology, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong Province, People's Republic of China
| | - Li Zhang
- Sun Yat-Sen University Cancer Center; State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong Province, People's Republic of China.,Department of Pediatric Oncology, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong Province, People's Republic of China
| | - Yanjie Xu
- Department of Hematology, Tianjin Medical University Cancer Institute and Hospital; National Clinical Research Center for Cancer; Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin, People's Republic of China
| | - Suying Lu
- Sun Yat-Sen University Cancer Center; State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong Province, People's Republic of China.,Department of Pediatric Oncology, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong Province, People's Republic of China
| | - Jia Zhu
- Sun Yat-Sen University Cancer Center; State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong Province, People's Republic of China.,Department of Pediatric Oncology, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong Province, People's Republic of China
| | - Junting Huang
- Sun Yat-Sen University Cancer Center; State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong Province, People's Republic of China.,Department of Pediatric Oncology, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong Province, People's Republic of China
| | - Juan Wang
- Sun Yat-Sen University Cancer Center; State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong Province, People's Republic of China.,Department of Pediatric Oncology, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong Province, People's Republic of China
| | - Yang Hu
- Department of Hematology, Tianjin Medical University Cancer Institute and Hospital; National Clinical Research Center for Cancer; Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin, People's Republic of China
| | - Yizhuo Zhang
- Department of Hematology, Tianjin Medical University Cancer Institute and Hospital; National Clinical Research Center for Cancer; Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin, People's Republic of China.,Sun Yat-Sen University Cancer Center; State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong Province, People's Republic of China.,Department of Pediatric Oncology, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong Province, People's Republic of China
| |
Collapse
|
83
|
Functional interplay among thiol-based redox signaling, metabolism, and ferroptosis unveiled by a genetic variant of TP53. Proc Natl Acad Sci U S A 2020; 117:26804-26811. [PMID: 33055209 DOI: 10.1073/pnas.2009943117] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The p53 tumor suppressor protein is a transcription factor and master stress response mediator, and it is subject to reduction-oxidation (redox)-dependent regulation. The P47S variant of TP53, which exists primarily in African-descent populations, associates with an elevated abundance of low molecular weight (LMW) thiols, including glutathione (GSH) and coenzyme A (CoA). Here we show that S47 and P47 cells exhibit distinct metabolic profiles, controlled by their different redox states and expression of Activating Transcription Factor-4 (ATF4). We find that S47 cells exhibit decreased catabolic glycolysis but increased use of the pentose phosphate pathway (PPP), and an enhanced abundance of the antioxidant, NADPH. We identify ATF4 as differentially expressed in P47 and S47 cells and show that ATF4 can reverse the redox status and rescue metabolism of S47 cells, as well as increase sensitivity to ferroptosis. This adaptive metabolic switch is rapid, reversible, and accompanied by thiol-mediated changes in the structures and activities of key glycolytic signaling pathway proteins, including GAPDH and G6PD. The results presented here unveil the important functional interplay among pathways regulating thiol-redox status, metabolic adaptation, and cellular responses to oxidative stress.
Collapse
|
84
|
Pavez Lorie E, Stricker N, Plitta-Michalak B, Chen IP, Volkmer B, Greinert R, Jauch A, Boukamp P, Rapp A. Characterisation of the novel spontaneously immortalized and invasively growing human skin keratinocyte line HaSKpw. Sci Rep 2020; 10:15196. [PMID: 32938951 PMCID: PMC7494900 DOI: 10.1038/s41598-020-71315-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Accepted: 08/10/2020] [Indexed: 12/16/2022] Open
Abstract
We here present the spontaneously immortalised cell line, HaSKpw, as a novel model for the multistep process of skin carcinogenesis. HaSKpw cells were established from the epidermis of normal human adult skin that, without crisis, are now growing unrestricted and feeder-independent. At passage 22, clonal populations were established and clone7 (HaSKpwC7) was further compared to the also spontaneously immortalized HaCaT cells. As important differences, the HaSKpw cells express wild-type p53, remain pseudodiploid, and show a unique chromosomal profile with numerous complex aberrations involving chromosome 20. In addition, HaSKpw cells overexpress a pattern of genes and miRNAs such as KRT34, LOX, S100A9, miR21, and miR155; all pointing to a tumorigenic status. In concordance, HaSKpw cells exhibit reduced desmosomal contacts that provide them with increased motility and a highly migratory/invasive phenotype as demonstrated in scratch- and Boyden chamber assays. In 3D organotypic cultures, both HaCaT and HaSKpw cells form disorganized epithelia but only the HaSKpw cells show tumorcell-like invasive growth. Together, HaSKpwC7 and HaCaT cells represent two spontaneous (non-genetically engineered) “premalignant” keratinocyte lines from adult human skin that display different stages of the multistep process of skin carcinogenesis and thus represent unique models for analysing skin cancer development and progression.
Collapse
Affiliation(s)
- Elizabeth Pavez Lorie
- Leibniz Research Institute for Environmental Medicine, Auf'm Hennekamp 50, 40225, Düsseldorf, Germany
| | - Nicola Stricker
- Cell Biology and Epigenetics, Department of Biology, Technical University of Darmstadt, Schnittspahnstr. 10, 64287, Darmstadt, Germany
| | - Beata Plitta-Michalak
- Cell Biology and Epigenetics, Department of Biology, Technical University of Darmstadt, Schnittspahnstr. 10, 64287, Darmstadt, Germany
| | - I-Peng Chen
- Centre of Dermatology, Elbe Clinics, Am Krankenhaus 1, Buxtehude, 21614, Germany
| | - Beate Volkmer
- Centre of Dermatology, Elbe Clinics, Am Krankenhaus 1, Buxtehude, 21614, Germany
| | - Rüdiger Greinert
- Centre of Dermatology, Elbe Clinics, Am Krankenhaus 1, Buxtehude, 21614, Germany
| | - Anna Jauch
- Institute of Human Genetics, University Heidelberg, 69120, Heidelberg, Germany
| | - Petra Boukamp
- Leibniz Research Institute for Environmental Medicine, Auf'm Hennekamp 50, 40225, Düsseldorf, Germany.
| | - Alexander Rapp
- Cell Biology and Epigenetics, Department of Biology, Technical University of Darmstadt, Schnittspahnstr. 10, 64287, Darmstadt, Germany.
| |
Collapse
|
85
|
Campos A, Pereira R, Vaz A, Caetano T, Malta M, Oliveira J, Carvalho FP, Mendo S, Lourenço J. Metals and low dose IR: Molecular effects of combined exposures using HepG2 cells as a biological model. JOURNAL OF HAZARDOUS MATERIALS 2020; 396:122634. [PMID: 32304850 DOI: 10.1016/j.jhazmat.2020.122634] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 03/19/2020] [Accepted: 03/31/2020] [Indexed: 06/11/2023]
Abstract
Uranium mining sites produce residues rich in metals and radionuclides, that may contaminate all environmental matrices, exposing human and non-human biota to low doses of ionizing radiation (LDIR) and to the chemical toxicity of several metals. To date, experimental and radio-epidemiological studies do not provide conclusive evidence of LDIR induced cancer. However, co-exposures (LDIR plus other contaminants), may increase the risks. To determine the potential for genotoxic effects in human cells induced by the exposure to LDIR plus metals, HEPG2 cells were exposed to different concentrations of a uranium mine effluent for 96 h. DNA damage was evaluated using the comet assay and changes in the expression of tumor suppressor and oncogenes were determined using qPCR. Results show that effluent concentrations higher than 5%, induce significant DNA damage. Also, a significant under-expression of ATM and TP53 genes and a significant overexpression of GADD45a gene was observed. Results show that the exposure to complex mixtures cannot be disregarded, as effects were detected at very low doses. This study highlights the need for further studies to clarify the risks of exposure to LDIR along with other stressors, to fully review the IR exposure risk limits established for human and non-human biota.
Collapse
Affiliation(s)
- A Campos
- ICBAS & Department of Biology, Faculty of Sciences of the University of Porto, Rua do Campo Alegre s/n, 4169-007, Porto, Portugal
| | - R Pereira
- ICBAS & Department of Biology, Faculty of Sciences of the University of Porto, Rua do Campo Alegre s/n, 4169-007, Porto, Portugal; GreenUPorto- Sustainable Agrifood Production Research Centre, Rua do Campo Alegre s/n, 4169-007, Porto, Portugal.
| | - A Vaz
- Department of Biology & CESAM, University of Aveiro, Campus Universitário de Santiago, 3810-193, Aveiro, Portugal
| | - T Caetano
- Department of Biology & CESAM, University of Aveiro, Campus Universitário de Santiago, 3810-193, Aveiro, Portugal.
| | - M Malta
- Instituto Superior Técnico/Laboratório de Proteccão e Segurança Radiológica, Universidade de Lisboa, Estrada Nacional 10, Km 139, 2695-066 Bobadela LRS, Portugal.
| | - J Oliveira
- Instituto Superior Técnico/Laboratório de Proteccão e Segurança Radiológica, Universidade de Lisboa, Estrada Nacional 10, Km 139, 2695-066 Bobadela LRS, Portugal.
| | - F P Carvalho
- Instituto Superior Técnico/Laboratório de Proteccão e Segurança Radiológica, Universidade de Lisboa, Estrada Nacional 10, Km 139, 2695-066 Bobadela LRS, Portugal.
| | - S Mendo
- Department of Biology & CESAM, University of Aveiro, Campus Universitário de Santiago, 3810-193, Aveiro, Portugal.
| | - J Lourenço
- Department of Biology & CESAM, University of Aveiro, Campus Universitário de Santiago, 3810-193, Aveiro, Portugal.
| |
Collapse
|
86
|
Intronic TP53 Polymorphisms Are Associated with Increased Δ133TP53 Transcript, Immune Infiltration and Cancer Risk. Cancers (Basel) 2020; 12:cancers12092472. [PMID: 32882831 PMCID: PMC7563340 DOI: 10.3390/cancers12092472] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 08/26/2020] [Accepted: 08/28/2020] [Indexed: 12/30/2022] Open
Abstract
We investigated the influence of selected TP53 SNPs in exon 4 and intron 4 on cancer risk, clinicopathological features and expression of TP53 isoforms. The intron 4 SNPs were significantly over-represented in cohorts of mixed cancers compared to three ethnically matched controls, suggesting they confer increased cancer risk. Further analysis showed that heterozygosity at rs1042522(GC) and either of the two intronic SNPs rs9895829(TC) and rs2909430(AG) confer a 2.34-5.35-fold greater risk of developing cancer. These SNP combinations were found to be associated with shorter patient survival for glioblastoma and prostate cancer. Additionally, these SNPs were associated with tumor-promoting inflammation as evidenced by high levels of infiltrating immune cells and expression of the Δ133TP53 and TP53β transcripts. We propose that these SNP combinations allow increased expression of the Δ133p53 isoforms to promote the recruitment of immune cells that create an immunosuppressive environment leading to cancer progression.
Collapse
|
87
|
Abstract
Myosins constitute a superfamily of actin-based molecular motor proteins that mediates a variety of cellular activities including muscle contraction, cell migration, intracellular transport, the formation of membrane projections, cell adhesion, and cell signaling. The 12 myosin classes that are expressed in humans share sequence similarities especially in the N-terminal motor domain; however, their enzymatic activities, regulation, ability to dimerize, binding partners, and cellular functions differ. It is becoming increasingly apparent that defects in myosins are associated with diseases including cardiomyopathies, colitis, glomerulosclerosis, neurological defects, cancer, blindness, and deafness. Here, we review the current state of knowledge regarding myosins and disease.
Collapse
|
88
|
Chen PH, Tseng WHS, Chi JT. The Intersection of DNA Damage Response and Ferroptosis-A Rationale for Combination Therapeutics. BIOLOGY 2020; 9:E187. [PMID: 32718025 PMCID: PMC7464484 DOI: 10.3390/biology9080187] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 07/16/2020] [Accepted: 07/17/2020] [Indexed: 12/17/2022]
Abstract
Ferroptosis is a novel form of iron-dependent cell death characterized by lipid peroxidation. While the importance and disease relevance of ferroptosis are gaining recognition, much remains unknown about its interaction with other biological processes and pathways. Recently, several studies have identified intricate and complicated interplay between ferroptosis, ionizing radiation (IR), ATM (ataxia-telangiectasia mutated)/ATR (ATM and Rad3-related), and tumor suppressor p53, which signifies the participation of the DNA damage response (DDR) in iron-related cell death. DDR is an evolutionarily conserved response triggered by various DNA insults to attenuate proliferation, enable DNA repairs, and dispose of cells with damaged DNA to maintain genome integrity. Deficiency in proper DDR in many genetic disorders or tumors also highlights the importance of this pathway. In this review, we will focus on the biological crosstalk between DDR and ferroptosis, which is mediated mostly via noncanonical mechanisms. For clinical applications, we also discuss the potential of combining ionizing radiation and ferroptosis-inducers for synergistic effects. At last, various ATM/ATR inhibitors under clinical development may protect ferroptosis and treat many ferroptosis-related diseases to prevent cell death, delay disease progression, and improve clinical outcomes.
Collapse
Affiliation(s)
- Po-Han Chen
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710, USA; (P.-H.C.); (W.H.-S.T.)
- Center for Genomic and Computational Biology, Duke University School of Medicine, Durham, NC 27710, USA
- Department of Molecular, Cellular, and Developmental Biology, Yale University, New Haven, CT 06511, USA
| | - Watson Hua-Sheng Tseng
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710, USA; (P.-H.C.); (W.H.-S.T.)
- Center for Genomic and Computational Biology, Duke University School of Medicine, Durham, NC 27710, USA
- Institute of Clinical Medicine, National Yang-Ming University, Taipei 112, Taiwan
| | - Jen-Tsan Chi
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710, USA; (P.-H.C.); (W.H.-S.T.)
- Center for Genomic and Computational Biology, Duke University School of Medicine, Durham, NC 27710, USA
| |
Collapse
|
89
|
Sambyal V, Kaur S, Manjari M, Uppal MS, Singh NR, Sudan M, Guleria K. Linkage disequilibrium and haplotypes of five TP53 polymorphisms in oesophageal cancer patients. J Genet 2020. [DOI: 10.1007/s12041-020-01224-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
90
|
Lieschke E, Wang Z, Kelly GL, Strasser A. Discussion of some 'knowns' and some 'unknowns' about the tumour suppressor p53. J Mol Cell Biol 2020; 11:212-223. [PMID: 30496435 PMCID: PMC6478126 DOI: 10.1093/jmcb/mjy077] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Revised: 10/22/2018] [Accepted: 11/27/2018] [Indexed: 12/13/2022] Open
Abstract
Activation of the tumour suppressor p53 upon cellular stress can induce a number of different cellular processes. The diverse actions of these processes are critical for the protective function of p53 in preventing the development of cancer. However, it is still not fully understood which process(es) activated by p53 is/are critical for tumour suppression and how this might differ depending on the type of cells undergoing neoplastic transformation and the nature of the drivers of oncogenesis. Moreover, it is not clear why upon activation of p53 some cells undergo cell cycle arrest and senescence whereas others die by apoptosis. Here we discuss some of the cellular processes that are crucial for p53-mediated tumour suppression and the factors that could impact cell fate upon p53 activation. Finally, we describe therapies aimed either at activating wild-type p53 or at changing the behaviour of mutant p53 to unleash tumour growth suppressive processes for therapeutic benefit in malignant disease.
Collapse
Affiliation(s)
- Elizabeth Lieschke
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, Australia.,Department of Medical Biology, University of Melbourne, Melbourne, Australia
| | - Zilu Wang
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, Australia.,Department of Medical Biology, University of Melbourne, Melbourne, Australia
| | - Gemma L Kelly
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, Australia.,Department of Medical Biology, University of Melbourne, Melbourne, Australia
| | - Andreas Strasser
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, Australia.,Department of Medical Biology, University of Melbourne, Melbourne, Australia
| |
Collapse
|
91
|
Hyun DH. Insights into the New Cancer Therapy through Redox Homeostasis and Metabolic Shifts. Cancers (Basel) 2020; 12:cancers12071822. [PMID: 32645959 PMCID: PMC7408991 DOI: 10.3390/cancers12071822] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 07/02/2020] [Accepted: 07/06/2020] [Indexed: 12/18/2022] Open
Abstract
Modest levels of reactive oxygen species (ROS) are necessary for intracellular signaling, cell division, and enzyme activation. These ROS are later eliminated by the body’s antioxidant defense system. High amounts of ROS cause carcinogenesis by altering the signaling pathways associated with metabolism, proliferation, metastasis, and cell survival. Cancer cells exhibit enhanced ATP production and high ROS levels, which allow them to maintain elevated proliferation through metabolic reprograming. In order to prevent further ROS generation, cancer cells rely on more glycolysis to produce ATP and on the pentose phosphate pathway to provide NADPH. Pro-oxidant therapy can induce more ROS generation beyond the physiologic thresholds in cancer cells. Alternatively, antioxidant therapy can protect normal cells by activating cell survival signaling cascades, such as the nuclear factor erythroid 2-related factor 2 (Nrf2)-Kelch-like ECH-associated protein 1 (Keap1) pathway, in response to radio- and chemotherapeutic drugs. Nrf2 is a key regulator that protects cells from oxidative stress. Under normal conditions, Nrf2 is tightly bound to Keap1 and is ubiquitinated and degraded by the proteasome. However, under oxidative stress, or when treated with Nrf2 activators, Nrf2 is liberated from the Nrf2-Keap1 complex, translocated into the nucleus, and bound to the antioxidant response element in association with other factors. This cascade results in the expression of detoxifying enzymes, including NADH-quinone oxidoreductase 1 (NQO1) and heme oxygenase 1. NQO1 and cytochrome b5 reductase can neutralize ROS in the plasma membrane and induce a high NAD+/NADH ratio, which then activates SIRT1 and mitochondrial bioenergetics. NQO1 can also stabilize the tumor suppressor p53. Given their roles in cancer pathogenesis, redox homeostasis and the metabolic shift from glycolysis to oxidative phosphorylation (through activation of Nrf2 and NQO1) seem to be good targets for cancer therapy. Therefore, Nrf2 modulation and NQO1 stimulation could be important therapeutic targets for cancer prevention and treatment.
Collapse
Affiliation(s)
- Dong-Hoon Hyun
- Department of Life Science, Ewha Womans University, Seoul 03760, Korea
| |
Collapse
|
92
|
Mehta A, Vasudevan S, Sharma SK, Panigrahi M, Suryavanshi M, Saifi M, Batra U. Biomarker testing for advanced lung cancer by next-generation sequencing; a valid method to achieve a comprehensive glimpse at mutational landscape. ACTA ACUST UNITED AC 2020. [DOI: 10.1186/s41241-020-00089-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Abstract
Background
Next-generation sequencing (NGS) based assay for finding an actionable driver in non-small-cell lung cancer is a less used modality in clinical practice. With a long list of actionable targets, limited tissue, arduous single-gene assays, the alternative of NGS for broad testing in one experiment looks attractive. We report here our experience with NGS for biomarker testing in hundred advanced lung cancer patients.
Methods
Predictive biomarker testing was performed using the Ion AmpliSeq™ Cancer Hotspot Panel V2 (30 tumors) and Oncomine™ Solid Tumor DNA and Oncomine™ Solid Tumor Fusion Transcript kit (70 tumors) on Ion-Torrent sequencing platform.
Results
One-seventeen distinct aberrations were detected across 29 genes in eighty-six tumors. The most commonly mutated genes were TP53 (43% cases), EGFR (23% cases) and KRAS (17% cases). Thirty-four patients presented an actionable genetic variant for which targeted therapy is presently available, and fifty-two cases harbored non-actionable variants with the possibility of recruitment in clinical trials. NGS results were validated by individual tests for detecting EGFR mutation, ALK1 rearrangement, ROS1 fusion, and c-MET amplification. Compared to single test, NGS exhibited good agreement for detecting EGFR mutations and ALK1 fusion (sensitivity- 88.89%, specificity- 100%, Kappa-score 0.92 and sensitivity- 80%, specificity- 100%, Kappa-score 0.88; respectively). Further, the response of patients harboring tyrosine kinase inhibitor (TKI) sensitizing EGFR mutations was assessed. The progression-free-survival of EGFR positive patients on TKI therapy, harboring a concomitant mutation in PIK3CA-mTOR and/or RAS-RAF-MAPK pathway gene and/or TP53 gene was inferior to those with sole-sensitizing EGFR mutation (2 months vs. 9.5 months, P = 0.015).
Conclusions
This is the first study from South Asia looking into the analytical validity of NGS and describing the mutational landscape of lung cancer patients to study the impact of co-mutations on cancer biology and treatment outcome. Our study demonstrates the clinical utility of NGS testing for identifying actionable variants and making treatment decisions in advanced lung cancer.
Collapse
|
93
|
Heidari Z, Harati‐Sadegh M, Arian A, Maruei‐Milan R, Salimi S. The effect of
TP53
and
P21
gene polymorphisms on papillary thyroid carcinoma susceptibility and clinical/pathological features. IUBMB Life 2020; 72:922-930. [DOI: 10.1002/iub.2225] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Accepted: 12/20/2019] [Indexed: 12/12/2022]
Affiliation(s)
- Zahra Heidari
- Department of EndocrinologySchool of Medicine, Zahedan University of Medical Sciences Zahedan Iran
| | - Mahdiyeh Harati‐Sadegh
- Genetic of Non‐Communicable Disease Research CenterZahedan University of Medical Sciences Zahedan Iran
| | - Abtin Arian
- Department of RadiologySchool of Medicine, Zahedan University of Medical Sciences Zahedan Iran
| | - Rostam Maruei‐Milan
- Department of Clinical BiochemistrySchool of Medicine, Zahedan University of Medical Sciences Zahedan Iran
| | - Saeedeh Salimi
- Department of Clinical BiochemistrySchool of Medicine, Zahedan University of Medical Sciences Zahedan Iran
- Cellular and Molecular Research Center, Resistant Tuberculosis InstituteZahedan University of Medical Sciences Zahedan Iran
| |
Collapse
|
94
|
Kamiza AB, Kamiza S, Singini MG, Mathew CG. Association of TP53 rs1042522 with cervical cancer in the sub-Saharan African population: a meta-analysis. Trop Med Int Health 2020; 25:666-672. [PMID: 32233050 DOI: 10.1111/tmi.13397] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
OBJECTIVE TP53 plays a crucial role in preventing cancer development. Previous studies in sub-Saharan Africa (SSA) reported inconclusive findings for the association of the TP53 rs1042522 C > G variant with cervical cancer. We therefore performed a meta-analysis to summarise this association in the SSA population. METHODS Online databases were searched to identify suitable articles according to the PRISMA guidelines. We included studies published in English or French that provided the sample sizes and genotype counts for both cases and controls and evaluated the association between TP53 rs1042522 and cervical cancer in the SSA population. A fixed-effect model was used to calculate the pooled odds ratio (OR) and 95% confidence intervals (95% CIs). RESULTS A total of 699 cervical cancer cases and 1008 controls from eight studies in SSA were included in this meta-analysis. Women harbouring the variant G allele of the TP53 rs1042522 were at increased risk of cervical cancer in allelic (G vs. C; OR = 1.30, 95% Cl = 1.12-1.50), homozygous (GG vs. CC; OR = 1.62, 95% CI = 1.20-2.19) and recessive (GG vs. CG + GG; OR = 1.74, 95% CI = 1.34-2.25) genetic models. However, the dominant genetic model (CG + GG vs. CC; OR = 1.20, 95% CI = 0.96-1.48) was not significantly associated with cervical cancer. CONCLUSIONS Our meta-analysis revealed that harbouring variant G allele of TP53 rs1042522 is associated with cervical cancer risk in the SSA population.
Collapse
Affiliation(s)
- Abram Bunya Kamiza
- Sydney Brenner Institute for Molecular Bioscience, University of the Witwatersrand, Johannesburg, South Africa
| | - Steve Kamiza
- Histopathology Department, College of Medicine, University of Malawi, Blantyre, Malawi
| | - Mwiza Gideon Singini
- National Cancer Registry, National Health Laboratory Service, Johannesburg, South Africa.,School of Public Health, University of the Witwatersrand, Johannesburg, South Africa
| | - Christopher George Mathew
- Sydney Brenner Institute for Molecular Bioscience, University of the Witwatersrand, Johannesburg, South Africa.,Department of Medical and Molecular Genetics, King's College London, London, UK
| |
Collapse
|
95
|
An update on the central nervous system manifestations of Li-Fraumeni syndrome. Acta Neuropathol 2020; 139:669-687. [PMID: 31468188 DOI: 10.1007/s00401-019-02055-3] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Revised: 08/01/2019] [Accepted: 08/02/2019] [Indexed: 12/19/2022]
Abstract
Li-Fraumeni syndrome (LFS), caused by the germline mutations in the TP53 gene, leads to significant lifetime risk to cancer in the central nervous system. Recognition of LFS, and elucidating its underlying cause has had a remarkable effect on our knowledge of the biology of brain tumors and represents a significant opportunity for cancer surveillance and screening. In this review, we discuss the historical context of the LFS with an emphasis on the clinicopathologic implications in clincal diagnosis, germline testing, and clinical management of brain tumor patients.
Collapse
|
96
|
Icen-Taskin I, Irtegun-Kandemir S, Munzuroglu O. TP53 rs1042522 polymorphism and early-onset breast cancer. JOURNAL OF RESEARCH IN MEDICAL SCIENCES 2020; 25:25. [PMID: 32419782 PMCID: PMC7213006 DOI: 10.4103/jrms.jrms_506_19] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 10/21/2019] [Accepted: 02/18/2020] [Indexed: 02/04/2023]
Abstract
Background: Breast cancer is the leading cause of cancer deaths among women. Early-onset breast cancer is well recognized as it clinically differs from old-age diagnosed breast neoplasms. TP53 rs1042522 polymorphism relates to the risk of breast neoplasms, but this relationship in Turkish early-onset breast cancer patients has not been investigated yet. We aimed to search the relationship between TP53 rs1042522 polymorphism and young Turkish breast cancer patients. Materials and Methods: Ninety-six female breast cancer patients who were ≤ 40 years of age and 96 healthy controls were enrolled in our study. Participants were genotyped by the hybridization probe system. Results: We identified that the genotype frequencies of rs1042522 were significantly different between controls and cases (P = 0.027). Participants carrying CG genotype had also reduced breast cancer risk (odds ratio = 0.4196, 95% confidence interval: 0.1941–0.9067, P = 0.027). Our results revealed that there is an association between GG and CG + CC genotype groups with progesterone receptor (PgR) status (P = 0.0219). Conclusion: Our findings indicate that the CG genotype is a protective factor against breast neoplasms. No other clinicopathologic parameters except for PgR status were found to be related to rs1042522 polymorphism in young Turkish breast cancer patients.
Collapse
Affiliation(s)
- Irmak Icen-Taskin
- Department of Molecular Biology and Genetics, Faculty of Science and Art, Inonu University, Malatya, Turkey
| | - Sevgi Irtegun-Kandemir
- Department of Medical Biology, Faculty of Medicine, Dicle University, Diyarbakir, Turkey
| | - Omer Munzuroglu
- Department of Bioengineering, Faculty of Engineering, Firat University, Elazig, Turkey
| |
Collapse
|
97
|
Abstract
Lung cancer in women is a modern epidemic and a major health crisis. Cigarette smoking remains the most important risk factor for lung cancer, and unfortunately smoking rates are either stabilized or continue to increase among women. Women may not be more susceptible to the carcinogenic effects of tobacco, but the biology of lung cancer differs between the sexes. This paper summarizes the biological sex differences in lung cancer, including molecular abnormalities, growth factor receptors, hormonal influences, DNA repair capacity, as well as differences in the histology and treatment outcomes of lung cancer in women.
Collapse
Affiliation(s)
- Christina R MacRosty
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Interventional Pulmonary Program, University of North Carolina at Chapel Hill, 130 Mason Farm Road, Chapel Hill, NC 27599-7020, USA
| | - M Patricia Rivera
- Division on Pulmonary and Critical Medicine, University of North Carolina at Chapel Hill, 130 Mason Farm Road, Suite 4125, Chapel Hill, NC 27599-7020, USA.
| |
Collapse
|
98
|
Deepa, Pundir S, Pundir C. Detection of tumor suppressor protein p53 with special emphasis on biosensors: A review. Anal Biochem 2020; 588:113473. [PMID: 31610154 DOI: 10.1016/j.ab.2019.113473] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 10/07/2019] [Accepted: 10/08/2019] [Indexed: 01/21/2023]
|
99
|
Thomas F, Giraudeau M, Renaud F, Ujvari B, Roche B, Pujol P, Raymond M, Lemaitre JF, Alvergne A. Can postfertile life stages evolve as an anticancer mechanism? PLoS Biol 2019; 17:e3000565. [PMID: 31805037 PMCID: PMC6917346 DOI: 10.1371/journal.pbio.3000565] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Revised: 12/17/2019] [Indexed: 12/14/2022] Open
Abstract
Why a postfertile stage has evolved in females of some species has puzzled evolutionary biologists for over 50 years. We propose that existing adaptive explanations have underestimated in their formulation an important parameter operating both at the specific and the individual levels: the balance between cancer risks and cancer defenses. During their life, most multicellular organisms naturally accumulate oncogenic processes in their body. In parallel, reproduction, notably the pregnancy process in mammals, exacerbates the progression of existing tumors in females. When, for various ecological or evolutionary reasons, anticancer defenses are too weak, given cancer risk, older females could not pursue their reproduction without triggering fatal metastatic cancers, nor even maintain a normal reproductive physiology if the latter also promotes the growth of existing oncogenic processes, e.g., hormone-dependent malignancies. At least until stronger anticancer defenses are selected for in these species, females could achieve higher inclusive fitness by ceasing their reproduction and/or going through menopause (assuming that these traits are easier to select than anticancer defenses), thereby limiting the risk of premature death due to metastatic cancers. Because relatively few species experience such an evolutionary mismatch between anticancer defenses and cancer risks, the evolution of prolonged life after reproduction could also be a rare, potentially transient, anticancer adaptation in the animal kingdom.
Collapse
Affiliation(s)
- Frédéric Thomas
- Centre de Recherches Ecologiques et Evolutives sur le Cancer/Centre de Recherches en Ecologie et Evolution de la Santé, Unité Mixte de Recherches, Institut de Recherches pour le Développement 224-Centre National de la Recherche Scientifique 5290-Université de Montpellier, Montpellier, France
| | - Mathieu Giraudeau
- Centre de Recherches Ecologiques et Evolutives sur le Cancer/Centre de Recherches en Ecologie et Evolution de la Santé, Unité Mixte de Recherches, Institut de Recherches pour le Développement 224-Centre National de la Recherche Scientifique 5290-Université de Montpellier, Montpellier, France
| | - François Renaud
- Centre de Recherches Ecologiques et Evolutives sur le Cancer/Centre de Recherches en Ecologie et Evolution de la Santé, Unité Mixte de Recherches, Institut de Recherches pour le Développement 224-Centre National de la Recherche Scientifique 5290-Université de Montpellier, Montpellier, France
| | - Beata Ujvari
- Centre for Integrative Ecology, School of Life and Environmental Sciences, Deakin University, Waurn Ponds, Victoria, Australia
- School of Natural Sciences, University of Tasmania, Hobart, Tasmania, Australia
| | - Benjamin Roche
- Centre de Recherches Ecologiques et Evolutives sur le Cancer/Centre de Recherches en Ecologie et Evolution de la Santé, Unité Mixte de Recherches, Institut de Recherches pour le Développement 224-Centre National de la Recherche Scientifique 5290-Université de Montpellier, Montpellier, France
- Unité mixte internationale de Modélisation Mathématique et Informatique des Systèmes Complexes, Unité Mixte de Recherches, Institut de Recherches pour le développement/Sorbonne Université, France
- Departamento de Etología, Fauna Silvestre y Animales de Laboratorio, Facultad de Medicina Veterinaria y Zootecnia, Universidad Nacional Autónoma de México (UNAM), Ciudad de México, México
| | - Pascal Pujol
- Centre de Recherches Ecologiques et Evolutives sur le Cancer/Centre de Recherches en Ecologie et Evolution de la Santé, Unité Mixte de Recherches, Institut de Recherches pour le Développement 224-Centre National de la Recherche Scientifique 5290-Université de Montpellier, Montpellier, France
- CHU Arnaud de Villeneuve, Montpellier, France
| | - Michel Raymond
- ISEM, Université de Montpellier, CNRS, IRD, EPHE, Montpellier, France
| | - Jean-François Lemaitre
- Centre National de la Recherche Scientifique, Unité mixte de recherche 5558, Laboratoire de Biométrie et Biologie Evolutive, Université Lyon 1 Villeurbanne, France
| | - Alexandra Alvergne
- ISEM, Université de Montpellier, CNRS, IRD, EPHE, Montpellier, France
- Institute of Social and Cultural Anthropology, School of Anthropology and Museum Ethnography, University of Oxford, United Kingdom
| |
Collapse
|
100
|
Liu X, Ren Z, Xu Y, Sun W, Li Y, Rui X, Xie D, Meng X, Zheng Z. Establishment and characterization of novel human primary endometrial cancer cell line (ZJB-ENC1) and its genomic characteristic. J Cancer 2019; 10:6466-6474. [PMID: 31772679 PMCID: PMC6856739 DOI: 10.7150/jca.33013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Accepted: 08/26/2019] [Indexed: 12/12/2022] Open
Abstract
The establishment of human malignant tumor cell lines can provide abundant experimental materials for understanding the biological characteristics of tumors, studying the carcinogenesis, molecular genetics and the mechanism of metastasis and evolution. In this study, a novel cell line designated ZJB-ENC1 has been established from poorly differentiated endometrioid adenocarcinoma. Cytological results showed monolayer-cultured cells were polygonal in shape and a piling-up tendency without contact inhabitation. Immunohistochemistry analysis showed that the cells were negative for ER, PR, c-erbB2, E-CAD, CD117, and OCT3/4, but strongly positive for PTEN and P16. Meanwhile, the tumorigenicity of ZJB-ENC1 was confirmed by subcutaneous transplantation of the cells into a xenograft mouse model. In addition, the results of the whole exome sequencing revealed a unique genomic characteristic of ZJB-ENC1 cells, all common and novel SNPs and InDels were identified. In conclusion, this new stable cell line may promote basic and clinical research on endometrial cancer (EC).
Collapse
Affiliation(s)
- Xiaozhen Liu
- Institute of Cancer and Basic Medicine (ICBM), Chinese Academy of Sciences, Hangzhou, 310022, China.,The Experimental Center, Cancer Hospital of the University of Chinese Academy of Sciences, Hangzhou, 310022, China.,The Experimental Center, Zhejiang cancer hospital , Hangzhou, 310022, China
| | - Zhuozhuo Ren
- Medical Support Department, Zhejiang Provincial Hospital of Traditional Chinese Medicine, Hangzhou 310022, China
| | - Yu Xu
- Department of Hematology, The First Affiliated Hospital, Zhejiang University, School of Medicine, Hangzhou, 310022, China
| | - Wei Sun
- Institute of Cancer and Basic Medicine (ICBM), Chinese Academy of Sciences, Hangzhou, 310022, China.,The Experimental Center, Cancer Hospital of the University of Chinese Academy of Sciences, Hangzhou, 310022, China.,The Experimental Center, Zhejiang cancer hospital , Hangzhou, 310022, China
| | - Yongfeng Li
- Institute of Cancer and Basic Medicine (ICBM), Chinese Academy of Sciences, Hangzhou, 310022, China.,Department of Breast Surgery, Cancer Hospital of the University of Chinese Academy of Sciences, Hangzhou, 310022, China.,Department of Breast Surgery, Zhejiang cancer hospital, Hangzhou, 310022, China
| | - Xinmiao Rui
- The Second Clinical Department, Zhejiang Chinese Medical University, Hangzhou, 310022, China
| | - Dafei Xie
- General Surgery Department, Zhejiang Hospital, Hangzhou, 310022, China
| | - Xuli Meng
- Department of Breast Thyroid Surgery, Tongde Hospital of Zhejiang Province, Hangzhou, 310022, China
| | - Zhiguo Zheng
- Institute of Cancer and Basic Medicine (ICBM), Chinese Academy of Sciences, Hangzhou, 310022, China.,The Experimental Center, Cancer Hospital of the University of Chinese Academy of Sciences, Hangzhou, 310022, China.,The Experimental Center, Zhejiang cancer hospital , Hangzhou, 310022, China
| |
Collapse
|