1
|
Tong G, Shen Y, Li H, Qian H, Tan Z. NLRC4, inflammation and colorectal cancer (Review). Int J Oncol 2024; 65:99. [PMID: 39239759 PMCID: PMC11387119 DOI: 10.3892/ijo.2024.5687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 08/19/2024] [Indexed: 09/07/2024] Open
Abstract
Chronic inflammation is recognized as a major risk factor for cancer and is involved in every phase of the disease. Inflammasomes are central to the inflammatory response and play a crucial role in cancer development. The present review summarizes the role of Nod‑like receptor C4 (NLRC4) in inflammation and colorectal cancer (CRC). Reviews of the literature were conducted using Web of Science, PubMed and CNKI, with search terms including 'NLRC4', 'colorectal cancer', 'auto‑inflammatory diseases' and 'prognosis'. Variants of NLRC4 can cause recessive immune dysregulation and autoinflammation or lead to ulcerative colitis as a heterozygous risk factor. Additionally, genetic mutations in inflammasome components may increase susceptibility to cancer. NLRC4 is considered a tumor suppressor in CRC. The role of NLRC4 in CRC signaling pathways is currently understood to involve five key aspects (caspase 1, NLRP3/IL‑8, IL‑1β/IL‑1, NAIP and p53). The mechanisms by which NLRC4 is involved in CRC are considered to be threefold (through pyroptosis, apoptosis, necroptosis and PANoptosis; regulating the immune response; and protecting intestinal epithelial cells to prevent CRC). However, the impact of NLRC4 mutations on CRC remains unclear. In conclusion, NLRC4 is a significant inflammasome that protects against CRC through various signaling pathways and mechanisms. The association between NLRC4 mutations and CRC warrants further investigation.
Collapse
Affiliation(s)
- Guojun Tong
- Department of Colorectal Surgery, Huzhou Central Hospital, The Affiliated Central Hospital of Huzhou University, Huzhou, Zhejiang 313003, P.R. China
- Central Laboratory, Huzhou Central Hospital, The Affiliated Central Hospital of Huzhou University, Huzhou, Zhejiang 313003, P.R. China
| | - Yan Shen
- Department of General Surgery, Huzhou Central Hospital, The Affiliated Central Hospital of Huzhou University, Huzhou, Zhejiang 313003, P.R. China
| | - Hui Li
- Department of General Surgery, Huzhou Central Hospital, The Affiliated Central Hospital of Huzhou University, Huzhou, Zhejiang 313003, P.R. China
| | - Hai Qian
- Department of General Surgery, Huzhou Central Hospital, The Affiliated Central Hospital of Huzhou University, Huzhou, Zhejiang 313003, P.R. China
| | - Zhenhua Tan
- Department of General Surgery, Huzhou Central Hospital, The Affiliated Central Hospital of Huzhou University, Huzhou, Zhejiang 313003, P.R. China
| |
Collapse
|
2
|
Thawornkuno C, Srisuksai K, Simanon N, Adisakwattana P, Ampawong S, Boonyuen U, Limpanont Y, Chusongsang P, Chusongsang Y, Kiangkoo N, Reamtong O. A reanalysis and integration of transcriptomics and proteomics datasets unveil novel drug targets for Mekong schistosomiasis. Sci Rep 2024; 14:12969. [PMID: 38839835 PMCID: PMC11153569 DOI: 10.1038/s41598-024-63869-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 06/03/2024] [Indexed: 06/07/2024] Open
Abstract
Schistosomiasis, caused by Schistosoma trematodes, is a significant global health concern, particularly affecting millions in Africa and Southeast Asia. Despite efforts to combat it, the rise of praziquantel (PZQ) resistance underscores the need for new treatment options. Protein kinases (PKs) are vital in cellular signaling and offer potential as drug targets. This study focused on focal adhesion kinase (FAK) as a candidate for anti-schistosomal therapy. Transcriptomic and proteomic analyses of adult S. mekongi worms identified FAK as a promising target due to its upregulation and essential role in cellular processes. Molecular docking simulations assessed the binding energy of FAK inhibitors to Schistosoma FAK versus human FAK. FAK inhibitor 14 and PF-03814735 exhibited strong binding to Schistosoma FAK with minimal binding for human FAK. In vitro assays confirmed significant anti-parasitic activity against S. mekongi, S. mansoni, and S. japonicum, comparable to PZQ, with low toxicity in human cells, indicating potential safety. These findings highlight FAK as a promising target for novel anti-schistosomal therapies. However, further research, including in vivo studies, is necessary to validate efficacy and safety before clinical use. This study offers a hopeful strategy to combat schistosomiasis and reduce its global impact.
Collapse
Affiliation(s)
- Charin Thawornkuno
- Department of Molecular Tropical Medicine and Genetics, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Krittika Srisuksai
- Department of Molecular Tropical Medicine and Genetics, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Nattapon Simanon
- Department of Molecular Tropical Medicine and Genetics, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Poom Adisakwattana
- Department of Helminthology, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Sumate Ampawong
- Department of Tropical Pathology, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Usa Boonyuen
- Department of Molecular Tropical Medicine and Genetics, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Yanin Limpanont
- Department of Social and Environmental Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Phiraphol Chusongsang
- Department of Social and Environmental Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Yupa Chusongsang
- Department of Social and Environmental Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Nuttapohn Kiangkoo
- Department of Social and Environmental Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Onrapak Reamtong
- Department of Molecular Tropical Medicine and Genetics, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand.
| |
Collapse
|
3
|
Rocha VA, Aquino AM, Magosso N, Souza PV, Justulin LA, Domeniconi RF, Barbisan LF, Romualdo GR, Scarano WR. 2,4-dichlorophenoxyacetic acid (2,4-D) exposure during postnatal development alters the effects of western diet on mouse prostate. Reprod Toxicol 2023; 120:108449. [PMID: 37516258 DOI: 10.1016/j.reprotox.2023.108449] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 07/24/2023] [Accepted: 07/26/2023] [Indexed: 07/31/2023]
Abstract
Western diet (WD), abundant in saturated fats and simple carbohydrates, has been associated with the development of prostate diseases. In addition, 2,4-dichlorophenoxyacetic acid (2,4-D), an herbicide used in agricultural and non-agricultural settings, may interfere with the endocrine system impacting reproductive health. The association of both factors is something common in everyday life, however, there are no relevant studies associating them as possible modulators of prostatic diseases. This study evaluated the action of the herbicide 2,4-D on the postnatal development of the prostate in mice fed with WD. Male C57Bl/6J mice received simultaneously a WD and 2,4-D at doses of 0.02, 2.0, or 20.0 mg/kg b.w./day for 6 months. The prolongated WD intake induced obesity and glucose intolerance, increasing body weight and fat. WD induced morphological changes and increased PCNA-positive epithelial cells in prostate. Additionally, the WD increased gene expression of AR, antioxidant targets, inflammation-related cytokines, cell repair and turnover, and targets related to methylation and miRNAs biosynthesis compared to the counterpart (basal diet). 2,4-D (0.02 and 2.0) changed prostate morphology and gene expression evoked by WD. In contrast, the WD group exposed to 20 mg/kg of 2,4-D reduced feed intake and body weight, and increased expression of androgen receptor and genes related to cell repair and DNA methylation compared to the negative control. Our results showed that 2,4-D was able to modulate the effects caused by WD, mainly at lower doses. However, further studies are needed to elucidate the mechanisms of 2,4-D on the obesogenic environment caused by the WD.
Collapse
Affiliation(s)
- V A Rocha
- São Paulo State University (UNESP), Department of Structural and Functional Biology, Institute of Biosciences, Botucatu, São Paulo, Brazil
| | - A M Aquino
- São Paulo State University (UNESP), Department of Structural and Functional Biology, Institute of Biosciences, Botucatu, São Paulo, Brazil
| | - N Magosso
- São Paulo State University (UNESP), Department of Structural and Functional Biology, Institute of Biosciences, Botucatu, São Paulo, Brazil
| | - P V Souza
- São Paulo State University (UNESP), Department of Structural and Functional Biology, Institute of Biosciences, Botucatu, São Paulo, Brazil
| | - L A Justulin
- São Paulo State University (UNESP), Department of Structural and Functional Biology, Institute of Biosciences, Botucatu, São Paulo, Brazil
| | - R F Domeniconi
- São Paulo State University (UNESP), Department of Structural and Functional Biology, Institute of Biosciences, Botucatu, São Paulo, Brazil
| | - L F Barbisan
- São Paulo State University (UNESP), Department of Structural and Functional Biology, Institute of Biosciences, Botucatu, São Paulo, Brazil
| | - G R Romualdo
- São Paulo State University (UNESP), Department of Structural and Functional Biology, Institute of Biosciences, Botucatu, São Paulo, Brazil; São Paulo State University (UNESP), Botucatu Medical School, Experimental Research Unit (UNIPEX), Multimodel Drug Screening Platform - Laboratory of Chemically induced and Experimental Carcinogenesis (MDSP-LCQE), Botucatu, SP, Brazil
| | - W R Scarano
- São Paulo State University (UNESP), Department of Structural and Functional Biology, Institute of Biosciences, Botucatu, São Paulo, Brazil.
| |
Collapse
|
4
|
Chen P, Chen Y, Yan ZQ, Ding SY, Liu HP, Tu JQ, Zhang XW. Protective Effect of the Polysaccharides from Taraxacum mongolicum Leaf by Modulating the p53 Signaling Pathway in H22 Tumor-Bearing Mice. Foods 2022; 11:3340. [PMID: 36359953 PMCID: PMC9656931 DOI: 10.3390/foods11213340] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 10/13/2022] [Accepted: 10/18/2022] [Indexed: 09/14/2023] Open
Abstract
Dandelion is an edible plant with a variety of bioactive components. This paper mainly reports the antitumor activity of dandelion polysaccharide DLP120 on H22 tumor-bearing mice. DLP120 is an acidic polysaccharide composed of pectin and arabinogalactan. The results indicate that DLP120 markedly inhibited tumor growth in a dose-dependent manner and attenuated and regulated negative effects on organs. In addition, DLP120 not only increased the viability of spleen lymphocytes and natural killer (NK) cells, but also increased the proportion of lymphocyte subsets in peripheral blood. Furthermore, Hematoxylin-Eosin (HE) staining showed that tumor tissues and cells exhibited typical pathology features. Annexin V FITC/PI staining and cell cycle distribution results further confirmed apoptosis and cell cycle arrest in S and G2 phases. Notably, there was a significant accumulation of reactive oxygen species. Western blotting results demonstrated that the expression of p53 was up-regulated in the DLP120 group. Moreover, the pro-apoptotic protein Bax was up-regulated while the inhibitory-apoptotic protein Bcl-2 was down-regulated. In addition, the expression of Fas and FasL, associated with the death receptor pathway, were also up-regulated. Overall, administration of DLP120 in H22 tumor-bearing mice can not only enhance immunity but also directly induce tumor cell apoptosis.
Collapse
Affiliation(s)
| | | | | | | | - Hui-Ping Liu
- State Key Laboratory of Food Nutrition and Safety, Ministry of Education of China, College of Food Engineering and Biotechnology, Tianjin University of Science &Technology, Tianjin 300457, China
| | | | | |
Collapse
|
5
|
Zhang L, Hou N, Chen B, Kan C, Han F, Zhang J, Sun X. Post-Translational Modifications of p53 in Ferroptosis: Novel Pharmacological Targets for Cancer Therapy. Front Pharmacol 2022; 13:908772. [PMID: 35685623 PMCID: PMC9171069 DOI: 10.3389/fphar.2022.908772] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 05/10/2022] [Indexed: 12/21/2022] Open
Abstract
The tumor suppressor p53 is a well-known cellular guardian of genomic integrity that blocks cell cycle progression or induces apoptosis upon exposure to cellular stresses. However, it is unclear how the remaining activities of p53 are regulated after the abrogation of these routine activities. Ferroptosis is a form of iron- and lipid-peroxide-mediated cell death; it is particularly important in p53-mediated carcinogenesis and corresponding cancer prevention. Post-translational modifications have clear impacts on the tumor suppressor function of p53. Here, we review the roles of post-translational modifications in p53-mediated ferroptosis, which promotes the elimination of tumor cells. A thorough understanding of the p53 functional network will be extremely useful in future strategies to identify pharmacological targets for cancer therapy.
Collapse
Affiliation(s)
- Le Zhang
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, China
- Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Ningning Hou
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, China
- Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Bing Chen
- Department of Pathology, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Chengxia Kan
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, China
- Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Fang Han
- Department of Pathology, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Jingwen Zhang
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, China
- Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, China
- *Correspondence: Jingwen Zhang, ; Xiaodong Sun,
| | - Xiaodong Sun
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, China
- Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, China
- *Correspondence: Jingwen Zhang, ; Xiaodong Sun,
| |
Collapse
|
6
|
Xie Z, Lu G, Zhou R, Ma Y. Thiacloprid-induced hepatotoxicity in zebrafish: Activation of the extrinsic and intrinsic apoptosis pathways regulated by p53 signaling pathway. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2022; 246:106147. [PMID: 35349858 DOI: 10.1016/j.aquatox.2022.106147] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 03/03/2022] [Accepted: 03/20/2022] [Indexed: 05/21/2023]
Abstract
Thiacloprid (THCP) is one of the major neonicotinoid insecticides, and its wide use has led to high detection in various media of aquatic environment, posing potential risks to aquatic organisms. This study was focused on the phenotypic responses and mechanisms of toxicity in zebrafish (Danio rerio) upon treatment with waterborne THCP (0.4, 4 and 40 μM) for 21 days in vivo or 412.9 μM for 24 h in vitro. In vivo, we found that THCP induced severe oxidative stress, hepatic abnormalities, leakage of alanine aminotransferase and aspartate aminotransferase and apoptosis. The analysis of RNA-sequencing suggested the activation of the p53 signaling pathway under THCP exposure. The following in vitro study showed that THCP intoxication activated reactive oxygen species (ROS)-dependent p53 signaling pathway and induced hepatotoxicity in the zebrafish liver cells. The addition of p53 inhibitor pifithrin-α (10 μM) exerted protection against of THCP-induced hepatotoxicity by reducing oxidative stress and inhibiting the p53 signaling pathway and apoptosis. Moreover, gene expression analyses indicated that both the extrinsic and intrinsic apoptosis pathways were involved in apoptosis induced by p53 activation. Overall, our results suggest that activation of the p53 signaling pathway is an important mechanism of THCP-induced hepatotoxicity.
Collapse
Affiliation(s)
- Zhongtang Xie
- Key Laboratory of Integrated Regulation and Resources Development of Shallow Lakes of Ministry of Education, College of Environment, Hohai University, Nanjing 210098, PR China
| | - Guanghua Lu
- Key Laboratory of Integrated Regulation and Resources Development of Shallow Lakes of Ministry of Education, College of Environment, Hohai University, Nanjing 210098, PR China.
| | - Ranran Zhou
- Key Laboratory of Integrated Regulation and Resources Development of Shallow Lakes of Ministry of Education, College of Environment, Hohai University, Nanjing 210098, PR China
| | - Yuchen Ma
- Key Laboratory of Integrated Regulation and Resources Development of Shallow Lakes of Ministry of Education, College of Environment, Hohai University, Nanjing 210098, PR China
| |
Collapse
|
7
|
Zhang Y, Ma X, Li H, Zhuang J, Feng F, Liu L, Liu C, Sun C. Identifying the Effect of Ursolic Acid Against Triple-Negative Breast Cancer: Coupling Network Pharmacology With Experiments Verification. Front Pharmacol 2021; 12:685773. [PMID: 34858165 PMCID: PMC8631906 DOI: 10.3389/fphar.2021.685773] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 10/20/2021] [Indexed: 11/13/2022] Open
Abstract
Triple negative breast cancer (TNBC) is a subtype of breast cancer with complex heterogeneity, high invasiveness, and long-term poor prognosis. With the development of molecular pathology and molecular genetics, the gene map of TNBC with distinctive biological characteristics has been outlined more clearly. Natural plant extracts such as paclitaxel, vinblastine, colchicine etc., have occupied an important position in the treatment of hormone-independent breast cancer. Ursolic acid (UA), a triterpenoid acid compound derived from apple, pear, loquat leaves, etc., has been reported to be effective in a variety of cancer treatments, but there are few reports on the treatment of TNBC. This study performed comprehensive bioinformatics analysis and in vitro experiments to identify the effect of UA on TNBC treatment and its potential molecular mechanism. Our results showed that UA could not only reduce the proliferation, migration, and invasion in MDA-MB-231 and MDA-MB-468 cell lines with a dose-dependent manner but also induce cell cycle arrest and apoptosis. Meanwhile, we collected the gene expression data GSE45827 and GSE65194 from GEO for comparison between TNBC and normal cell type and obtained 724 DEGs. Subsequently, PLK1 and CCNB1 related to TNBC were screened as the key targets via topological analysis and molecular docking, and gene set enrichment analysis identified the key pathway as the p53 signaling pathway. In addition, quantitative real-time PCR and western blot verified the key genes were PLK1 and CCNB1. In vivo and in vitro experiments showed that UA could inhibit the growth of TNBC cells, and down-regulate the protein expression levels of PLK1 and CCNB1 by mediating p53 signaling pathway. These findings provide strong evidence for UA intervention in TNBC via multi-target therapy.
Collapse
Affiliation(s)
- Yubao Zhang
- School of Basic Medicine, Qingdao University, Qingdao, China
| | - Xiaoran Ma
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Huayao Li
- College of Basic Medical, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jing Zhuang
- Department of Oncology, Weifang Traditional Chinese Hospital, Weifang, China
| | - Fubin Feng
- Department of Oncology, Weifang Traditional Chinese Hospital, Weifang, China
| | - Lijuan Liu
- Department of Oncology, Weifang Traditional Chinese Hospital, Weifang, China
| | - Cun Liu
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Changgang Sun
- Department of Oncology, Weifang Traditional Chinese Hospital, Weifang, China.,Qingdao Academy of Chinese Medical Sciences, Shandong University of Traditional Chinese Medicine, Qingdao, China
| |
Collapse
|
8
|
Ma J, Hu X, Dai B, Wang Q, Wang H. Prediction of the mechanism of miRNAs in laryngeal squamous cell carcinoma based on the miRNA-mRNA regulatory network. PeerJ 2021; 9:e12075. [PMID: 34513340 PMCID: PMC8395572 DOI: 10.7717/peerj.12075] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 08/06/2021] [Indexed: 12/19/2022] Open
Abstract
In this study, a bioinformatics analysis is conducted to screen differentially expressed miRNAs and mRNAs in laryngeal squamous cell carcinoma (LSCC). Based on this information, we explored the possible roles of miRNAs in the pathogenesis of LSCC. The RNA-Seq data from 79 laryngeal cancer samples in the Gene Expression Omnibus (GEO) database were sorted. Differentially expressed miRNAs and mRNAs in LSCC are screened using the PERL programming language, and it was analysed by Gene Ontology (GO) and the Kyoto Encyclopedia of Genes and Genomes (KEGG). The miRNA-mRNA regulatory network of LSCC is constructed using Cytoscape software. Then, quantitative real-time PCR (QRT- PCR), Cell Counting Kit-8 (CCK8) and flow cytometry analysis we are used to further validate key miRNAs. We identified 99 differentially expressed miRNAs and 2,758 differentially expressed mRNAs in LSCC tissues from the GEO database. Four more important miRNAs displaying a high degree of connectivity are selected, these results suggest that they play an important role in the pathogenesis of LSCC. As shown in the present study, we identified specific miRNA-mRNA networks associated with the occurrence and development of LSCC through bioinformatics analysis. We found a miRNA molecule closely related to LSCC based on miRNA-mRNA network: miR-140-3p was down-regulated in LSCC. In addition, the potential antitumor effect of miR-140-3p in LSCC was verified in the experiment, and it was proved that overexpression of miR-140-3p could inhibit the proliferation of LSCC cells and promote cell apoptosis, suggesting that miR-140-3p may be a potential tumor marker in LSCC.
Collapse
Affiliation(s)
- Jinhua Ma
- Department of Otolaryngology, Cangzhou Central Hospital, Cangzhou, China
| | - Xiaodong Hu
- Department of Otolaryngology, Cangzhou Central Hospital, Cangzhou, China
| | - Baoqiang Dai
- Department of Otolaryngology, Cangzhou Central Hospital, Cangzhou, China
| | - Qiang Wang
- Department of Otolaryngology, Cangzhou Central Hospital, Cangzhou, China
| | - Hongqin Wang
- Department of Otolaryngology, Cangzhou Central Hospital, Cangzhou, China
| |
Collapse
|
9
|
Recent Advances in Signaling Pathways Comprehension as Carcinogenesis Triggers in Basal Cell Carcinoma. J Clin Med 2020; 9:jcm9093010. [PMID: 32961989 PMCID: PMC7565128 DOI: 10.3390/jcm9093010] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Revised: 09/07/2020] [Accepted: 09/16/2020] [Indexed: 12/14/2022] Open
Abstract
Basal cell carcinoma (BCC) is the most common malignant skin tumor. BCC displays a different behavior compared with other neoplasms, has a slow evolution, and metastasizes very rarely, but sometimes it causes an important local destruction. Chronic ultraviolet exposure along with genetic factors are the most important risk factors involved in BCC development. Mutations in the PTCH1 gene are associated with Gorlin syndrome, an autosomal dominant disorder characterized by the occurrence of multiple BCCs, but are also the most frequent mutations observed in sporadic BCCs. PTCH1 encodes for PTCH1 protein, the most important negative regulator of the Hedgehog (Hh) pathway. There are numerous studies confirming Hh pathway involvement in BCC pathogenesis. Although Hh pathway has been intensively investigated, it remains incompletely elucidated. Recent studies on BCC tumorigenesis have shown that in addition to Hh pathway, there are other signaling pathways involved in BCC development. In this review, we present recent advances in BCC carcinogenesis.
Collapse
|
10
|
Rubin JB, Lagas JS, Broestl L, Sponagel J, Rockwell N, Rhee G, Rosen SF, Chen S, Klein RS, Imoukhuede P, Luo J. Sex differences in cancer mechanisms. Biol Sex Differ 2020; 11:17. [PMID: 32295632 PMCID: PMC7161126 DOI: 10.1186/s13293-020-00291-x] [Citation(s) in RCA: 165] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Accepted: 03/18/2020] [Indexed: 02/07/2023] Open
Abstract
We now know that cancer is many different diseases, with great variation even within a single histological subtype. With the current emphasis on developing personalized approaches to cancer treatment, it is astonishing that we have not yet systematically incorporated the biology of sex differences into our paradigms for laboratory and clinical cancer research. While some sex differences in cancer arise through the actions of circulating sex hormones, other sex differences are independent of estrogen, testosterone, or progesterone levels. Instead, these differences are the result of sexual differentiation, a process that involves genetic and epigenetic mechanisms, in addition to acute sex hormone actions. Sexual differentiation begins with fertilization and continues beyond menopause. It affects virtually every body system, resulting in marked sex differences in such areas as growth, lifespan, metabolism, and immunity, all of which can impact on cancer progression, treatment response, and survival. These organismal level differences have correlates at the cellular level, and thus, males and females can fundamentally differ in their protections and vulnerabilities to cancer, from cellular transformation through all stages of progression, spread, and response to treatment. Our goal in this review is to cover some of the robust sex differences that exist in core cancer pathways and to make the case for inclusion of sex as a biological variable in all laboratory and clinical cancer research. We finish with a discussion of lab- and clinic-based experimental design that should be used when testing whether sex matters and the appropriate statistical models to apply in data analysis for rigorous evaluations of potential sex effects. It is our goal to facilitate the evaluation of sex differences in cancer in order to improve outcomes for all patients.
Collapse
Affiliation(s)
- Joshua B Rubin
- Department of Pediatrics, Washington University School of Medicine, 660 South Euclid Avenue, St Louis, MO, 63110, USA.
- Department of Neuroscience, Washington University School of Medicine, 660 South Euclid Avenue, St Louis, MO, 63110, USA.
| | - Joseph S Lagas
- Department of Pediatrics, Washington University School of Medicine, 660 South Euclid Avenue, St Louis, MO, 63110, USA
| | - Lauren Broestl
- Department of Pediatrics, Washington University School of Medicine, 660 South Euclid Avenue, St Louis, MO, 63110, USA
| | - Jasmin Sponagel
- Department of Pediatrics, Washington University School of Medicine, 660 South Euclid Avenue, St Louis, MO, 63110, USA
| | - Nathan Rockwell
- Department of Pediatrics, Washington University School of Medicine, 660 South Euclid Avenue, St Louis, MO, 63110, USA
| | - Gina Rhee
- Department of Pediatrics, Washington University School of Medicine, 660 South Euclid Avenue, St Louis, MO, 63110, USA
| | - Sarah F Rosen
- Department of Medicine, Washington University School of Medicine, 660 South Euclid Avenue, St Louis, MO, 63110, USA
| | - Si Chen
- Department of Biomedical Engineering, Washington University School of Medicine, 660 South Euclid Avenue, St Louis, MO, 63110, USA
| | - Robyn S Klein
- Department of Neuroscience, Washington University School of Medicine, 660 South Euclid Avenue, St Louis, MO, 63110, USA
- Department of Medicine, Washington University School of Medicine, 660 South Euclid Avenue, St Louis, MO, 63110, USA
| | - Princess Imoukhuede
- Department of Biomedical Engineering, Washington University School of Medicine, 660 South Euclid Avenue, St Louis, MO, 63110, USA
| | - Jingqin Luo
- Department of Surgery, Washington University School of Medicine, 660 South Euclid Avenue, St Louis, MO, 63110, USA
| |
Collapse
|
11
|
Yang LY, Greig NH, Tweedie D, Jung YJ, Chiang YH, Hoffer BJ, Miller JP, Chang KH, Wang JY. The p53 inactivators pifithrin-μ and pifithrin-α mitigate TBI-induced neuronal damage through regulation of oxidative stress, neuroinflammation, autophagy and mitophagy. Exp Neurol 2019; 324:113135. [PMID: 31778663 DOI: 10.1016/j.expneurol.2019.113135] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2019] [Revised: 10/20/2019] [Accepted: 11/24/2019] [Indexed: 01/06/2023]
Abstract
Traumatic brain injury (TBI) is one of the most common causes of death and disability worldwide. We investigated whether inhibition of p53 using pifithrin (PFT)-α or PFT-μ provides neuroprotective effects via p53 transcriptional dependent or -independent mechanisms, respectively. Sprague Dawley rats were subjected to controlled cortical impact TBI followed by the administration of PFTα or PFT-μ (2 mg/kg, i.v.) at 5 h after TBI. Brain contusion volume, as well as sensory and motor functions were evaluated at 24 h after TBI. TBI-induced impairments were mitigated by both PFT-α and PFT-μ. Fluoro-Jade C staining was used to label degenerating neurons within the TBI-induced cortical contusion region that, together with Annexin V positive neurons, were reduced by PFT-μ. Double immunofluorescence staining similarly demonstrated that PFT-μ significantly increased HO-1 positive neurons and mRNA expression in the cortical contusion region as well as decreased numbers of 4-hydroxynonenal (4HNE)-positive cells. Levels of mRNA encoding for p53, autophagy, mitophagy, anti-oxidant, anti-inflammatory related genes and proteins were measured by RT-qPCR and immunohistochemical staining, respectively. PFT-α, but not PFT-μ, significantly lowered p53 mRNA expression. Both PFT-α and PFT-μ lowered TBI-induced pro-inflammatory cytokines (IL-1β and IL-6) mRNA levels as well as TBI-induced autophagic marker localization (LC3 and p62). Finally, treatment with PFT-μ mitigated TBI-induced declines in mRNA levels of PINK-1 and SOD2. Our data suggest that both PFT-μ and PFT-α provide neuroprotective actions through regulation of oxidative stress, neuroinflammation, autophagy, and mitophagy mechanisms, and that PFT-μ, in particular, holds promise as a TBI treatment strategy.
Collapse
Affiliation(s)
- Ling-Yu Yang
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
| | - Nigel H Greig
- Drug Design & Development Section, Translational Gerontology Branch, Intramural Research Program National Institute on Aging, NIH, Baltimore, MD 21224, USA
| | - David Tweedie
- Drug Design & Development Section, Translational Gerontology Branch, Intramural Research Program National Institute on Aging, NIH, Baltimore, MD 21224, USA
| | - Yoo Jin Jung
- Drug Design & Development Section, Translational Gerontology Branch, Intramural Research Program National Institute on Aging, NIH, Baltimore, MD 21224, USA
| | - Yung-Hsiao Chiang
- Department of Neurosurgery, Taipei Medical University Hospital, Taipei 110, Taiwan; Neuroscience Research Center, Taipei Medical University, Taipei 110, Taiwan
| | - Barry J Hoffer
- Department of Neurological Surgery, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Jonathan P Miller
- Department of Neurological Surgery, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Ke-Hui Chang
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
| | - Jia-Yi Wang
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 110, Taiwan; Department of Neurosurgery, Taipei Medical University Hospital, Taipei 110, Taiwan; Neuroscience Research Center, Taipei Medical University, Taipei 110, Taiwan.
| |
Collapse
|
12
|
Shi SW, Li B, Dong Y, Ge Y, Qu X, Lu LG, Yuan YH, Li LJ, Li Y. In Vitro and Clinical Studies of Gene Therapy with Recombinant Human Adenovirus-p53 Injection for Malignant Melanoma. HUM GENE THER CL DEV 2019; 30:7-18. [PMID: 30618300 DOI: 10.1089/humc.2018.112] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Malignant melanoma is an aggressive tumor with high fatality rates and poor prognosis, mainly due to the lack of efficient treatment methods. The present study investigated the potential antitumor effects of recombinant adenovirus p53 (rAd-p53) on human malignant melanoma. The optimal viral titer on a human malignant melanoma (A-375) cell line was determined for the rAd-p53 treatment. The invasive abilities, apoptosis, variations in the cell cycle, and molecular expression levels of A-375 cells were detected after infection by rAd-p53. A tumor growth curve and hematoxylin and eosin staining were carried out for experiments in nude mice. Twenty-one patients with malignant melanoma were evaluated, including 12 cases without gene therapy and nine cases with rAd-p53 gene therapy. The overall survival rate and the median survival time were analyzed between the two groups of patients. When the multiplicity of infection was 100, the cells showed the best transfection efficiency. The invasive ability, apoptosis, cycle changes of the cells, and the expression of the p53, p21, and Bax genes and proteins were significantly changed in the experimental group. In nude mice, the tumor growth curve and the tumor size in the experimental group were significantly smaller than those of the control group. Hematoxylin and eosin staining revealed tumor metastasis in the blank group and the control group but not in the experimental group. Between the two groups of patients, the median survival of the gene therapy group (38 months) was greater than that of the group without gene therapy (27 months). In this study, high expression of the p53 gene could regulate the gene expression and reduce the invasive and metastatic abilities of the tumor cells. Furthermore, rAd-p53 effectively improved the survival of patients with malignant melanoma. Therefore, rAd-p53 may be a potential treatment method for human malignant melanoma.
Collapse
Affiliation(s)
- Shan-Wei Shi
- 1 State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases and Department of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University, Chengdu, P.R. China.,2 Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Hospital of Stomatology, Institute of Stomatological Research, Sun Yat-sen University, Guangzhou, Guangdong, P.R. China
| | - Bo Li
- 1 State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases and Department of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University, Chengdu, P.R. China
| | - Yang Dong
- 1 State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases and Department of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University, Chengdu, P.R. China
| | - Yang Ge
- 1 State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases and Department of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University, Chengdu, P.R. China
| | - Xing Qu
- 1 State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases and Department of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University, Chengdu, P.R. China
| | - Li-Guang Lu
- 1 State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases and Department of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University, Chengdu, P.R. China
| | - Yi-Hang Yuan
- 1 State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases and Department of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University, Chengdu, P.R. China
| | - Long-Jiang Li
- 1 State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases and Department of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University, Chengdu, P.R. China
| | - Yi Li
- 1 State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases and Department of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University, Chengdu, P.R. China
| |
Collapse
|
13
|
Coelho A, Nogueira A, Soares S, Assis J, Pereira D, Bravo I, Catarino R, Medeiros R. TP53 Arg72Pro polymorphism is associated with increased overall survival but not response to therapy in Portuguese/Caucasian patients with advanced cervical cancer. Oncol Lett 2018; 15:8165-8171. [PMID: 29731921 DOI: 10.3892/ol.2018.8354] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Accepted: 02/09/2018] [Indexed: 11/05/2022] Open
Abstract
Identification of mechanisms that influence the therapeutic response and survival in patients with cancer is important. It is known that the genetic variability of the host, including presence of genetic polymorphisms in genes involved in DNA damage response, serves a crucial role in the prognosis of these patients. The present hospital-based retrospective cohort study aimed to evaluate the influence of TP53 Arg72Pro (rs1042522) polymorphism in the clinical outcome of 260 Caucasian patients diagnosed with cervical cancer and treated with concomitant radiotherapy and chemotherapy. The polymorphism genotyping was assessed using allelic discrimination by quantiative polymerase chain reaction. The results indicate that the TP53 Arg72Pro polymorphism did not significantly impact the response to therapy (P=0.571) nor disease-free survival (P=0.081). However, the polymorphism did influence overall survival, as increased median survival time was observed for patients carrying Arg/Pro genotype when compared with patients with Arg/Arg and Pro/Pro genotypes (126 months vs. 111 months, respectively; P=0.047). To conclude, the present findings suggest that a pharmacogenomic profile based on the genetic background of patients, including the analysis of the TP53 genotypes, may individualize treatment nad assist in the selection of therapies that may improve clinical outcome and lower toxicity for the patients.
Collapse
Affiliation(s)
- Ana Coelho
- Molecular Oncology and Viral Pathology Group, Portuguese Oncology Institute of Porto-Research Center (CI-IPOP), Portuguese Oncology Institute of Porto (IPO-Porto), 4200-072 Porto, Portugal
| | - Augusto Nogueira
- Molecular Oncology and Viral Pathology Group, Portuguese Oncology Institute of Porto-Research Center (CI-IPOP), Portuguese Oncology Institute of Porto (IPO-Porto), 4200-072 Porto, Portugal.,FMUP, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
| | - Sílvia Soares
- Molecular Oncology and Viral Pathology Group, Portuguese Oncology Institute of Porto-Research Center (CI-IPOP), Portuguese Oncology Institute of Porto (IPO-Porto), 4200-072 Porto, Portugal
| | - Joana Assis
- Molecular Oncology and Viral Pathology Group, Portuguese Oncology Institute of Porto-Research Center (CI-IPOP), Portuguese Oncology Institute of Porto (IPO-Porto), 4200-072 Porto, Portugal.,FMUP, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
| | - Deolinda Pereira
- Molecular Oncology and Viral Pathology Group, Portuguese Oncology Institute of Porto-Research Center (CI-IPOP), Portuguese Oncology Institute of Porto (IPO-Porto), 4200-072 Porto, Portugal.,Oncology Department, Portuguese Oncology Institute of Porto, 4200-072 Porto, Portugal
| | - Isabel Bravo
- Medical Physics, Radiobiology and Radioprotection Group, Portuguese Oncology Institute of Porto-Research Center (CI-IPOP), Portuguese Oncology Institute of Porto, 4200-072 Porto, Portugal
| | - Raquel Catarino
- Molecular Oncology and Viral Pathology Group, Portuguese Oncology Institute of Porto-Research Center (CI-IPOP), Portuguese Oncology Institute of Porto (IPO-Porto), 4200-072 Porto, Portugal
| | - Rui Medeiros
- Molecular Oncology and Viral Pathology Group, Portuguese Oncology Institute of Porto-Research Center (CI-IPOP), Portuguese Oncology Institute of Porto (IPO-Porto), 4200-072 Porto, Portugal.,FMUP, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal.,CEBIMED, Faculty of Health Sciences of Fernando Pessoa University, 4249-004 Porto, Portugal.,Research Department, Portuguese League Against Cancer (NRNorte), 4200-172 Porto, Portugal
| |
Collapse
|
14
|
Grohmann T, Penke M, Petzold-Quinque S, Schuster S, Richter S, Kiess W, Garten A. Inhibition of NAMPT sensitizes MOLT4 leukemia cells for etoposide treatment through the SIRT2-p53 pathway. Leuk Res 2018; 69:39-46. [PMID: 29653431 DOI: 10.1016/j.leukres.2018.04.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Revised: 03/31/2018] [Accepted: 04/04/2018] [Indexed: 10/17/2022]
Abstract
NAMPT (Nicotinamide phosphoribosyltransferase) catalyses the rate-limiting step in the NAD biosynthesis from nicotinamide and thereby regulates the activity of NAD-dependent enzymes. Cancer cells are highly dependent on NAD for energy and DNA repair processes and are assumed to be more susceptible to an inhibition of NAD synthesis than non-transformed cells. We aimed to investigate whether or not inhibition of NAMPT with its specific inhibitor FK866 can sensitize leukemia cells for chemotherapeutic agents. NAMPT protein abundance, enzymatic activity and NAD concentrations were significantly higher in Jurkat and Molt-4 leukemia cell lines compared to normal peripheral blood mononuclear cells. Combination of etoposide and FK866 caused increased cell death in leukemia cell lines compared to etoposide alone. Etoposide decreased protein abundance of NAD-dependent deacetylases SIRTUIN1. After combining etoposide and FK866 treatment SIRTUIN2 was further decreased and accumulation and acetylation of the downstream target p53 was further enhanced in MOLT4 cells. Concomitantly, protein abundance of p21 and cleaved BAX was increased. Targeting NAMPT could be a novel therapeutic strategy to enhance the efficacy of chemotherapeutic agents such as etoposide against leukemia.
Collapse
Affiliation(s)
- Theresa Grohmann
- Hospital for Children & Adolescents, Center for Pediatric Research Leipzig, University of Leipzig, Germany
| | - Melanie Penke
- Hospital for Children & Adolescents, Center for Pediatric Research Leipzig, University of Leipzig, Germany
| | - Stefanie Petzold-Quinque
- Hospital for Children & Adolescents, Center for Pediatric Research Leipzig, University of Leipzig, Germany
| | - Susanne Schuster
- Hospital for Children & Adolescents, Center for Pediatric Research Leipzig, University of Leipzig, Germany
| | - Sandy Richter
- Hospital for Children & Adolescents, Center for Pediatric Research Leipzig, University of Leipzig, Germany
| | - Wieland Kiess
- Hospital for Children & Adolescents, Center for Pediatric Research Leipzig, University of Leipzig, Germany
| | - Antje Garten
- Hospital for Children & Adolescents, Center for Pediatric Research Leipzig, University of Leipzig, Germany; University of Birmingham, Institute of Metabolism and Systems Research (IMSR), Birmingham, UK.
| |
Collapse
|
15
|
Current-day precision oncology: from cancer prevention, screening, drug development, and treatment - have we fallen short of the promise? Curr Opin Oncol 2017; 28:441-6. [PMID: 27428363 DOI: 10.1097/cco.0000000000000318] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
PURPOSE OF REVIEW Precision oncology has been a strategy of prevention, screening, and treatment. Although much has been invested, have the results fallen so far short of the promise? The advancement of technology and research has opened new doors, yet a variety of pitfalls are present. This review presents the successes, failures, and opportunities of precision oncology in the current landscape. RECENT FINDINGS The use of targeted gene sequencing and the overwhelming results of superresponders have generated much excitement and support for precision oncology from the medical community. Despite notable successes, many challenges still pave the way of precision oncology: intratumoral heterogeneity, the need for serial biopsies, availability of treatments, target prioritization, ethical issues with germline incidental findings, medical education, clinical trial design, and costs. SUMMARY Precision oncology shows much potential through the use of next-generation sequencing and molecular advances, but does this potential warrant the investment? There are many obstacles on the way of this technology that should make us question if the investment (both monetary and man-hours) will live up to the promise. The review aims to not criticize this technology, but to give a realistic view of where we are, especially regarding cancer treatment and prevention.
Collapse
|
16
|
Alcoba DD, Schneider J, Arruda L, Martiny PB, Capp E, von Eye Corleta H, Brum IS. Brilliant cresyl blue staining does not present cytotoxic effects on human luteinized follicular cells, according to gene/protein expression, as well as to cytotoxicity tests. Reprod Biol 2017; 17:60-68. [PMID: 28063736 DOI: 10.1016/j.repbio.2016.12.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Revised: 11/20/2016] [Accepted: 12/19/2016] [Indexed: 12/16/2022]
Abstract
In vitro oocyte maturation presents many advantages and its success is related to the selection of fully grown oocytes. In animal models, staining of cumulus-oocyte complexes (COCs) with Brilliant Cresyl Blue (BCB) is widely used for this selection. However, a lack of information about the safety of BCB makes its applicability in humans questionable. Because granulosa and cumulus cells have a close relationship with the oocyte and taking into account that follicular cells are also exposed to BCB when this pre-selection method is applied, we aimed to evaluate the effects of BCB on human follicular cells exposed to BCB. Cytotoxicity tests (Sulforhodamine B and Neutral Red Uptake) and gene and protein expression of elements related to the cell cycle (BAX, BCL2, TP53 and CDKN1A), as well as to cell death and metabolism (GAPDH), glucose consumption, and estradiol and progesterone secretion, were examined in granulosa and cumulus cells with and without exposure to BCB. Regardless estradiol secretion and glucose consumption, all other evaluations presented similar results between control and treated (BCB) groups, independently of cell type. Therefore, our results demonstrate convincingly that BCB seems to be safe for use in humans and it should encourage future studies to evaluate the development of embryos derived from human oocytes selected by BCB staining.
Collapse
Affiliation(s)
- Diego Duarte Alcoba
- Institute of Health Sciences/Department of Physiology, Universidade Federal do Rio Grande do Sul, 90050-170, Porto Alegre, Brazil.
| | - Júlia Schneider
- Institute of Health Sciences/Department of Physiology, Universidade Federal do Rio Grande do Sul, 90050-170, Porto Alegre, Brazil
| | - Letícia Arruda
- Institute of Health Sciences/Department of Physiology, Universidade Federal do Rio Grande do Sul, 90050-170, Porto Alegre, Brazil
| | - Patrícia Borba Martiny
- Institute of Health Sciences/Department of Physiology, Universidade Federal do Rio Grande do Sul, 90050-170, Porto Alegre, Brazil
| | - Edison Capp
- Service of Gynecology and Obstetrics, Hospital de Clínicas de Porto Alegre, 90035-903, Porto Alegre, Brazil; Department of Gynecological Endocrinology and Reproductive Medicine, University Women's Hospital, 69120, Heidelberg, Germany
| | - Helena von Eye Corleta
- Institute of Health Sciences/Department of Physiology, Universidade Federal do Rio Grande do Sul, 90050-170, Porto Alegre, Brazil; Service of Gynecology and Obstetrics, Hospital de Clínicas de Porto Alegre, 90035-903, Porto Alegre, Brazil
| | - Ilma Simoni Brum
- Institute of Health Sciences/Department of Physiology, Universidade Federal do Rio Grande do Sul, 90050-170, Porto Alegre, Brazil
| |
Collapse
|
17
|
Chemical Variations on the p53 Reactivation Theme. Pharmaceuticals (Basel) 2016; 9:ph9020025. [PMID: 27187415 PMCID: PMC4932543 DOI: 10.3390/ph9020025] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Revised: 05/06/2016] [Accepted: 05/09/2016] [Indexed: 01/31/2023] Open
Abstract
Among the tumor suppressor genes, p53 is one of the most studied. It is widely regarded as the "guardian of the genome", playing a major role in carcinogenesis. In fact, direct inactivation of the TP53 gene occurs in more than 50% of malignancies, and in tumors that retain wild-type p53 status, its function is usually inactivated by overexpression of negative regulators (e.g., MDM2 and MDMX). Hence, restoring p53 function in cancer cells represents a valuable anticancer approach. In this review, we will present an updated overview of the most relevant small molecules developed to restore p53 function in cancer cells through inhibition of the p53-MDMs interaction, or direct targeting of wild-type p53 or mutated p53. In addition, optimization approaches used for the development of small molecules that have entered clinical trials will be presented.
Collapse
|
18
|
Ross JS, Wang K, Gay L, Otto GA, White E, Iwanik K, Palmer G, Yelensky R, Lipson DM, Chmielecki J, Erlich RL, Rankin AN, Ali SM, Elvin JA, Morosini D, Miller VA, Stephens PJ. Comprehensive Genomic Profiling of Carcinoma of Unknown Primary Site: New Routes to Targeted Therapies. JAMA Oncol 2016; 1:40-49. [PMID: 26182302 DOI: 10.1001/jamaoncol.2014.216] [Citation(s) in RCA: 167] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
IMPORTANCE For carcinoma of unknown primary site (CUP), determining the primary tumor site may be uninformative and often does not improve outcome. OBJECTIVE To discover opportunities for targeted therapies in patients with CUP not currently searched for in routine practice. DESIGN, SETTING, AND PARTICIPANTS Comprehensive genomic profiling on 200 CUP formalin-fixed paraffin-embedded specimens (mean, 756× coverage) using the hybrid-capture-based FoundationOne assay at academic and community oncology clinics. MAIN OUTCOMES AND MEASURES Presence of targetable genomic alterations (GAs) in CUP and responses to targeted therapies. RESULTS There were 125 adenocarcinomas of unknown primary site (ACUPs) and 75 carcinomas of unknown primary site without features of adenocarcinoma (non-ACUPs). At least 1 GA was found in 192 (96%) of CUP specimens, with a mean (SD) of 4.2 (2.8) GAs per tumor. The most frequent GAs were in TP53 (110 [55%]), KRAS (40 [20%]), CDKN2A (37 [19%]), MYC (23 [12%]), ARID1A (21 [11%]), MCL1 (19 [10%]), PIK3CA (17 [9%]), ERBB2 (16 [8%]), PTEN (14 [7%]), EGFR (12 [6%]), SMAD4 (13 [7%]), STK11 (13 [7%]), SMARCA4 (12 [6%]), RB1 (12 [6%]), RICTOR (12 [6%]), MLL2 (12 [6%]), BRAF (11 [6%]), and BRCA2 (11 [6%]). One or more potentially targetable GAs were identified in 169 of 200 (85%) CUP specimens. Mutations or amplifications of ERBB2 were more frequent in ACUPs (13 [10%]) than in non-ACUPs (3 [4%]). Alterations of EGFR (10 [8%] vs 2 [3%]) and BRAF (8 [6%] vs 3 [4%]) were more common in ACUPs than in non-ACUPs. Strikingly, clinically relevant alterations in the receptor tyrosine kinase (RTK)/Ras signaling pathway including alterations in ALK, ARAF, BRAF, EGFR, FGFR1, FGFR2, KIT, KRAS, MAP2K1, MET, NF1, NF2, NRAS, RAF1, RET, and ROS1 were found in 90 (72%) ACUPs but in only 29 (39%) non-ACUPs (P < .001). CONCLUSIONS AND RELEVANCE Almost all CUP samples harbored at least 1 clinically relevant GA with potential to influence and personalize therapy. The ACUP tumors were more frequently driven by GAs in the highly druggable RTK/Ras/mitogen-activated protein kinase (MAPK) signaling pathway than the non-ACUP tumors. Comprehensive genomic profiling can identify novel treatment paradigms to address the limited options and poor prognoses of patients with CUP.
Collapse
Affiliation(s)
- Jeffrey S Ross
- Foundation Medicine Inc, Cambridge, Massachusetts.,Department of Pathology and Laboratory Medicine, Albany Medical College, Albany, New York
| | - Kai Wang
- Foundation Medicine Inc, Cambridge, Massachusetts
| | - Laurie Gay
- Foundation Medicine Inc, Cambridge, Massachusetts
| | - Geoff A Otto
- Foundation Medicine Inc, Cambridge, Massachusetts
| | - Emily White
- Foundation Medicine Inc, Cambridge, Massachusetts
| | - Kiel Iwanik
- Foundation Medicine Inc, Cambridge, Massachusetts
| | - Gary Palmer
- Foundation Medicine Inc, Cambridge, Massachusetts
| | | | | | | | | | | | - Siraj M Ali
- Foundation Medicine Inc, Cambridge, Massachusetts
| | | | | | | | | |
Collapse
|
19
|
Chae YK, Chung SY, Davis AA, Carneiro BA, Chandra S, Kaplan J, Kalyan A, Giles FJ. Adenoid cystic carcinoma: current therapy and potential therapeutic advances based on genomic profiling. Oncotarget 2015; 6:37117-34. [PMID: 26359351 PMCID: PMC4741919 DOI: 10.18632/oncotarget.5076] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Accepted: 08/08/2015] [Indexed: 11/25/2022] Open
Abstract
Adenoid cystic carcinoma (ACC) is a rare cancer with high potential for recurrence and metastasis. Efficacy of current treatment options, particularly for advanced disease, is very limited. Recent whole genome and exome sequencing has dramatically improved our understanding of ACC pathogenesis. A balanced translocation resulting in the MYB-NFIB fusion gene appears to be a fundamental signature of ACC. In addition, sequencing has identified a number of other driver genes mutated in downstream pathways common to other well-studied cancers. Overexpression of oncogenic proteins involved in cell growth, adhesion, cell cycle regulation, and angiogenesis are also present in ACC. Collectively, studies have identified genes and proteins for targeted, mechanism-based, therapies based on tumor phenotypes, as opposed to nonspecific cytotoxic agents. In addition, although few studies in ACC currently exist, immunotherapy may also hold promise. Better genetic understanding will enable treatment with novel targeted agents and initial exploration of immune-based therapies with the goal of improving outcomes for patients with ACC.
Collapse
Affiliation(s)
- Young Kwang Chae
- Northwestern Medicine Developmental Therapeutics Institute, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Su Yun Chung
- Northwestern Medicine Developmental Therapeutics Institute, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Andrew A. Davis
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Benedito A. Carneiro
- Northwestern Medicine Developmental Therapeutics Institute, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Sunandana Chandra
- Northwestern Medicine Developmental Therapeutics Institute, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Jason Kaplan
- Northwestern Medicine Developmental Therapeutics Institute, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Aparna Kalyan
- Northwestern Medicine Developmental Therapeutics Institute, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Francis J. Giles
- Northwestern Medicine Developmental Therapeutics Institute, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| |
Collapse
|
20
|
Jamil S, Lam I, Majd M, Tsai SH, Duronio V. Etoposide induces cell death via mitochondrial-dependent actions of p53. Cancer Cell Int 2015; 15:79. [PMID: 26251638 PMCID: PMC4527242 DOI: 10.1186/s12935-015-0231-z] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Accepted: 07/21/2015] [Indexed: 12/19/2022] Open
Abstract
Background Etoposide has been used clinically in cancer treatment, as well as in numerous research studies, for many years. However, there is incomplete information about its exact mechanism of action in induction of cell death. Methods Etoposide was compared at various concentrations to characterize the mechanisms by which it induces cell death. We investigated its effects on mouse embryonic fibroblasts (MEFs) and focused on both transcriptional and non-transcriptional responses of p53. Results Here we demonstrate that treatment of MEFs with higher concentrations of etoposide induce apoptosis and activate the transcription-dependent functions of p53. Interestingly, lower concentrations of etoposide also induced apoptosis, but without any evidence of p53-dependent transcription up-regulation. Treatment of MEFs with an inhibitor of p53, Pifithrin-α, blocked p53-dependent transcription but failed to rescue the cells from etoposide-induced apoptosis. Treatment with PES, which inhibits the mitochondrial arm of the p53 pathway inhibited etoposide-induced cell death at all concentrations tested. Conclusions We have demonstrated that transcriptional functions of p53 are dispensable for etoposide-induced cell death. The more recently characterized effects of p53 at the mitochondria, likely involving its interactions with BCL-2 family members, are thus more important for etoposide’s actions. Electronic supplementary material The online version of this article (doi:10.1186/s12935-015-0231-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Sarwat Jamil
- Department of Medicine, Jack Bell Research Centre, Vancouver Coastal Health Research Institute, University of British Columbia, 2660 Oak St., Vancouver, BC V6H 3Z6 Canada
| | - Irene Lam
- Department of Medicine, Jack Bell Research Centre, Vancouver Coastal Health Research Institute, University of British Columbia, 2660 Oak St., Vancouver, BC V6H 3Z6 Canada
| | - Maryam Majd
- Department of Medicine, Jack Bell Research Centre, Vancouver Coastal Health Research Institute, University of British Columbia, 2660 Oak St., Vancouver, BC V6H 3Z6 Canada
| | - Shu-Huei Tsai
- Department of Medicine, Jack Bell Research Centre, Vancouver Coastal Health Research Institute, University of British Columbia, 2660 Oak St., Vancouver, BC V6H 3Z6 Canada
| | - Vincent Duronio
- Department of Medicine, Jack Bell Research Centre, Vancouver Coastal Health Research Institute, University of British Columbia, 2660 Oak St., Vancouver, BC V6H 3Z6 Canada
| |
Collapse
|
21
|
Xu J, Acharya S, Sahin O, Zhang Q, Saito Y, Yao J, Wang H, Li P, Zhang L, Lowery FJ, Kuo WL, Xiao Y, Ensor J, Sahin AA, Zhang XHF, Hung MC, Zhang JD, Yu D. 14-3-3ζ turns TGF-β's function from tumor suppressor to metastasis promoter in breast cancer by contextual changes of Smad partners from p53 to Gli2. Cancer Cell 2015; 27:177-92. [PMID: 25670079 PMCID: PMC4325275 DOI: 10.1016/j.ccell.2014.11.025] [Citation(s) in RCA: 137] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2014] [Revised: 09/10/2014] [Accepted: 11/24/2014] [Indexed: 12/01/2022]
Abstract
Transforming growth factor β (TGF-β) functions as a tumor suppressor in premalignant cells but as a metastasis promoter in cancer cells. The dichotomous functions of TGF-β are proposed to be dictated by different partners of its downstream effector Smads. However, the mechanism for the contextual changes of Smad partners remained undefined. Here, we demonstrate that 14-3-3ζ destabilizes p53, a Smad partner in premalignant mammary epithelial cells, by downregulating 14-3-3σ, thus turning off TGF-β's tumor suppression function. Conversely, 14-3-3ζ stabilizes Gli2 in breast cancer cells, and Gli2 partners with Smads to activate PTHrP and promote TGF-β-induced bone metastasis. The 14-3-3ζ-driven contextual changes of Smad partners from p53 to Gli2 may serve as biomarkers and therapeutic targets of TGF-β-mediated cancer progression.
Collapse
Affiliation(s)
- Jia Xu
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Sunil Acharya
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; The University of Texas Graduate School of Biomedical Sciences, Houston, TX 77030, USA
| | - Ozgur Sahin
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Qingling Zhang
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Yohei Saito
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jun Yao
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Hai Wang
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Ping Li
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Lin Zhang
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; The University of Texas Graduate School of Biomedical Sciences, Houston, TX 77030, USA
| | - Frank J Lowery
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; The University of Texas Graduate School of Biomedical Sciences, Houston, TX 77030, USA
| | - Wen-Ling Kuo
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Yi Xiao
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Joe Ensor
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Aysegul A Sahin
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Xiang H-F Zhang
- Lester and Sue Smith Breast Center and Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Mien-Chie Hung
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; The University of Texas Graduate School of Biomedical Sciences, Houston, TX 77030, USA; Center for Molecular Medicine and Graduate Institute of Cancer Biology, China Medical University, Taichung 404, Taiwan
| | - Jitao David Zhang
- Pharmaceutical Research and Early Development, F. Hoffmann-La Roche, Ltd., 4070 Basel, Switzerland
| | - Dihua Yu
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; The University of Texas Graduate School of Biomedical Sciences, Houston, TX 77030, USA.
| |
Collapse
|
22
|
Chen DY, Dai DF, Hua Y, Qi WQ. p53 suppresses 14-3-3γ by stimulating proteasome-mediated 14-3-3γ protein degradation. Int J Oncol 2014; 46:818-24. [PMID: 25384678 DOI: 10.3892/ijo.2014.2740] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2014] [Accepted: 09/26/2014] [Indexed: 11/05/2022] Open
Abstract
14-3-3 proteins are a family of highly conserved polypeptides that interact with a large number of proteins and play a role in a wide variety of cellular processes. 14-3-3 proteins have been demonstrated overexpressed in several cancers and serving as potential oncogenes. In a previous study we showed one isoform of the 14-3-3 family, 14-3-3γ was negatively regulated by p53 through binding to its promoter and inhibiting its transcription. In the present study we investigated both p53 and 14-3-3γ protein levels in human lung cancerous tissues and normal lung tissues. We found 14-3-3γ expression correlated to p53 overexpression in lung cancer tissues. Ecotopic expression of wild-type p53, but not mutant p53 (R175H) suppressed both endogenous and exogenous 14-3-3γ in colon and lung cancer cell lines. Further examination demonstrated that p53 interacted with C-terminal domain of 14-3-3γ and induced 14-3-3γ ubiquitination. MG132, a specific inhibitor of the 26S proteasome, could block the effect of p53 on 14-3-3γ protein levels, suggesting that p53 suppressed 14-3-3γ by stimulating the process of proteasome-mediated degradation of 14-3-3γ. These results indicate that the inhibitory effect of p53 on 14-3-3γ is mediated also by a post-transcriptional mechanism. Loss of p53 function may result in upregulation of 14-3-3γ in lung cancers.
Collapse
Affiliation(s)
- De-Yu Chen
- Institute of Oncology, The Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu 212001, P.R. China
| | - Dong-Fang Dai
- Institute of Oncology, The Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu 212001, P.R. China
| | - Ye Hua
- Institute of Oncology, The Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu 212001, P.R. China
| | - Wen-Qing Qi
- Institute of Oncology, The Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu 212001, P.R. China
| |
Collapse
|
23
|
Silveira AL, Faheina-Martins GV, Maia RC, Araújo DAM. Compound A398, a novel podophyllotoxin analogue: cytotoxicity and induction of apoptosis in human leukemia cells. PLoS One 2014; 9:e107404. [PMID: 25221997 PMCID: PMC4164611 DOI: 10.1371/journal.pone.0107404] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2014] [Accepted: 08/15/2014] [Indexed: 12/25/2022] Open
Abstract
Despite advances in oncology research, cancer is one of the leading causes of death worldwide. Thus, there is a demand for the development of more selective and effective antitumor agents. This study showed that A398, a novel podophyllotoxin analogue, was cytotoxic to the HT-29, MCF-7, MOLT-4 and HL-60 tumor cell lines, being less active in human peripheral blood mononuclear cells and normal cell lines FGH and IEC-6. Tests using the HepG2 lineage indicated that its metabolites do not contribute to its cytotoxicity. In the HL-60 cells, A398 induced apoptosis in a time and concentration-dependent manner, promoting mitochondrial depolarization, inhibition of Bcl-2, phosphatidylserine exposure, activation of caspases -8, -9 and -3, and DNA fragmentation. The production of reactive oxygen species does not seem to be a crucial event for the apoptotic process. Pretreatment with specific inhibitors of kinases ERK1/2, JNK and p38 resulted in an increased percentage of death induced by A398. These results indicate that the compound induced apoptosis through activation of intrinsic and extrinsic death pathways with the mechanism involving the inhibition of the MAPKs and Bcl-2. Taken together, our findings suggest that A398 has an anticancer potential, proving itself to be a candidate for preclinical studies.
Collapse
Affiliation(s)
- Alethéia L. Silveira
- Departamento de Biotecnologia, Universidade Federal da Paraíba, João Pessoa, Paraíba, Brasil
| | - Glaúcia V. Faheina-Martins
- Departamento de Biotecnologia, Universidade Federal da Paraíba, João Pessoa, Paraíba, Brasil
- Departamento de Biologia Molecular, Universidade Federal da Paraíba, João Pessoa, Paraíba, Brasil
| | - Raquel C. Maia
- Laboratório de Hemato-Oncologia Celular e Molecular, Instituto Nacional de Câncer (INCA), Rio de Janeiro, Rio de Janeiro, Brasil
| | - Demetrius A. M. Araújo
- Departamento de Biotecnologia, Universidade Federal da Paraíba, João Pessoa, Paraíba, Brasil
- * E-mail:
| |
Collapse
|
24
|
Ning XH, Zhang N, Li T, Wu PJ, Wang X, Li XY, Peng SH, Wang JY, Chen JC, Gong K. Telomere shortening is associated with genetic anticipation in Chinese Von Hippel-Lindau disease families. Cancer Res 2014; 74:3802-9. [PMID: 24986515 DOI: 10.1158/0008-5472.can-14-0024] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Von Hippel-Lindau (VHL) disease is a rare autosomal dominant cancer syndrome. A phenomenon known as genetic anticipation has been documented in some hereditary cancer syndromes, where it was proved to relate to telomere shortening. Because studies of this phenomenon in VHL disease have been relatively scarce, we investigated anticipation in 18 Chinese VHL disease families. We recruited 34 parent-child patient pairs (57 patients) from 18 families with VHL disease. Onset age was defined as the age when any symptom or sign of VHL disease first appeared. Anticipation of onset age was analyzed by paired t test and the other two special tests (HV and RY2). Relative telomere length of peripheral leukocytes was measured in 29 patients and 325 healthy controls. Onset age was younger in child than in parent in 31 of the 34 parent-child pairs. Patients in the first generation had older onset age with longer age-adjusted relative telomere length, and those in the next generation had younger onset age with shorter age-adjusted relative telomere length (P < 0.001) in the 10 parent-child pairs from eight families with VHL disease. In addition, relative telomere length was shorter in the 29 patients with VHL disease than in the normal controls (P = 0.003). The anticipation may relate to the shortening of telomere length in patients with VHL in successive generations. These findings indicate that anticipation is present in families with VHL disease and may be helpful for genetic counseling for families with VHL disease families and for further understanding the pathogenesis of VHL disease.
Collapse
Affiliation(s)
- Xiang-Hui Ning
- Authors' Affiliations: Departments of Urology and Institute of Urology, Peking University; National Urological Cancer Center
| | - Ning Zhang
- Department of Urology, Beijing Chaoyang Hospital, Capital University of Medicine Science, Beijing, P.R. China
| | - Teng Li
- Authors' Affiliations: Departments of Urology and Institute of Urology, Peking University; National Urological Cancer Center
| | - Peng-Jie Wu
- Authors' Affiliations: Departments of Urology and Institute of Urology, Peking University; National Urological Cancer Center
| | - Xi Wang
- Authors' Affiliations: Departments of Urology and Institute of Urology, Peking University; National Urological Cancer Center
| | - Xue-Ying Li
- Medical Statistics, Peking University First Hospital
| | - Shuang-He Peng
- Authors' Affiliations: Departments of Urology and Institute of Urology, Peking University; National Urological Cancer Center
| | - Jiang-Yi Wang
- Authors' Affiliations: Departments of Urology and Institute of Urology, Peking University; National Urological Cancer Center
| | - Jin-Chao Chen
- Authors' Affiliations: Departments of Urology and Institute of Urology, Peking University; National Urological Cancer Center
| | - Kan Gong
- Authors' Affiliations: Departments of Urology and Institute of Urology, Peking University; National Urological Cancer Center;
| |
Collapse
|
25
|
Ju W, Zhang M, Petrus M, Maeda M, Pise-Masison CA, Waldmann TA. Combination of 9-aminoacridine with Campath-1H provides effective therapy for a murine model of adult T-cell leukemia. Retrovirology 2014; 11:43. [PMID: 24890041 PMCID: PMC4060757 DOI: 10.1186/1742-4690-11-43] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2014] [Accepted: 05/15/2014] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Adult T-cell leukemia/lymphoma (ATL) is an aggressive malignancy of CD4+CD25+ lymphocytes caused by human T-cell lymphotropic virus type 1. While much progress has been made in understanding the mechanisms of cellular dysregulation, the prognosis for aggressive ATL still remains poor. Therefore, new therapeutic approaches need to be developed. RESULTS Previously, we demonstrated that the viral protein Tax inactivates p53 in HTLV-1-infected T-cells. Here we show that 9-aminoacridine (9AA) through p53 reactivation and NF-κB inhibition has selective toxicity for infected leukemic cells independent of their p53 status. We further demonstrate that 9AA activates caspase-3/7 resulting in PARP cleavage. Next we investigated the efficacy of 9AA in the MET-1 ATL model. Alone, 9AA did not cause significant drops in surrogate tumor markers, soluble IL-2Rα or β2-micorglobulin (β2μ) levels with only a slight increase in survival of MET-1-bearing mice. However, in combination with Campath-1H, 9AA treatment resulted in low soluble IL-2Rα and β2μ levels at 2 and 4 weeks. Consistent with reduced tumor cell burden, combination treatment significantly increased survival of MET-1-bearing mice compared to mice treated with either drug alone. Splenic cells isolated from 9AA or combination treated mice showed increased p53 protein levels and transcriptional activity. Consistent with increased tumor suppressor activity, we found increased PARP-1 cleavage in 9AA and combination treated cells. CONCLUSION Our results indicate that targeting reactivation of p53 and inhibition of NF-κB with acridine-derivatives in combination with other chemotherapeutics could result in increased efficacy and selective killing of tumor cells.
Collapse
Affiliation(s)
| | | | | | | | - Cynthia A Pise-Masison
- Lymphoid Malignancies Branch, Center for Cancer Research, National Cancer Institute, NIH, 10 Center Drive, Building 10, Room 4 N115, Bethesda, MD 20892-1374, USA.
| | | |
Collapse
|
26
|
Inhibition of ERN1 modifies the hypoxic regulation of the expression of TP53-related genes in U87 glioma cells. ENDOPLASMIC RETICULUM STRESS IN DISEASES 2014. [DOI: 10.2478/ersc-2014-0001] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
AbstractInhibition of ERN1 (endoplasmic reticulum to nuclei 1), the major signalling pathway of endoplasmic reticulum stress, significantly decreases tumor growth. We have studied the expression of tumor protein 53 (TP53)- related genes such as TOPORS (topoisomerase I binding, arginine/serine-rich, E3 ubiquitin protein ligase), TP53BP1 (TP53 binding protein 1), TP53BP2, SESN1 (sestrin 1), NME6 (non-metastatic cells 6), and ZMAT3 (zinc finger, Matrin-type 3) in glioma cells expressing dominantnegative ERN1 under baseline and hypoxic conditions. We demonstrated that inhibition of ERN1 function in U87 glioma cells resulted in increased expression of RYBP, TP53BP2, and SESN1 genes, but decreased expression of TP53BP1, TOPORS, NME6, and ZMAT3 genes. Moreover, inhibition of ERN1 affected hypoxia-mediated changes in expression of TP53-related genes and their magnitude. Indeed, hypoxia has no effect on expression of TP53BP1 and SESN1 in control cells, while resulted in increased expression of these genes in cells with inhibited ERN1 function. Magnitude of hypoxia-mediated changes in expression levels of RYBP and TP53BP2 was gene specific and more robust in the case of TP53BP2. Hypoxiamediated decrease in expression levels of TOPORS was more prominent if ERN1 was inhibited. Present study demonstrates that fine-tuning of the expression of TP53- associated genes depends upon endoplasmic reticulum stress signaling under normal and hypoxic conditions. Inhibition of ERN1 branch of endoplasmic reticulum stress response correlates with deregulation of p53 signaling and slower tumor growth.
Collapse
|