1
|
Hanuman Singh D, Deeksha W, Rajakumara E. Characterization of PARP1 binding to c-KIT1 G-quadruplex DNA: Insights into domain-specific interactions. Biophys Chem 2024; 315:107330. [PMID: 39342702 DOI: 10.1016/j.bpc.2024.107330] [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: 06/02/2024] [Revised: 09/10/2024] [Accepted: 09/24/2024] [Indexed: 10/01/2024]
Abstract
Poly(ADP-ribose) polymerase 1 (PARP1) is a nuclear enzyme involved in catalyzing Poly-(ADP-ribosyl)ation. PARP1 binds to different forms of DNA and DNA breaks and thus plays important roles in several cellular processes, including DNA damage repair, cell cycle regulation, chromatin remodeling, and maintaining genomic stability. In this study, we conducted biochemical and biophysical characterization of PARP1 binding to G-quadruplex DNA (G4-DNA). Our investigation identified ZnF1, ZnF3, and WGR as the critical domains to mediate PARP1 binding to G4-c-KIT1. Also, our results show that these domains together show cooperativity for G4-c-KIT1 recognition. Further, we establish that the presence of an oxidized (5-carboxylcytosine) base in the loop region of G4-c-KIT1 (G4-5caC-cKIT1) does not affect its recognition by PARP1. Both G4-c-KIT1 and G4-5caC-cKIT1 are potent stimulators of PARP1's catalytic activity. Our study advances the understanding of PARP1's versatile DNA binding capabilities for G4-c-KIT1 DNA irrespective of the oxidation/ modification in the DNA base. These insights into PARP1's domain-specific contributions to G4-c-KIT1 DNA recognition and catalysis expand our knowledge of its multifaceted roles in DNA repair and genome maintenance.
Collapse
Affiliation(s)
- Dagur Hanuman Singh
- Macromolecular Structural Biology Lab, Department of Biotechnology, Indian Institute of Technology Hyderabad, Kandi, Sangareddy, Telangana 502284, India
| | - Waghela Deeksha
- Macromolecular Structural Biology Lab, Department of Biotechnology, Indian Institute of Technology Hyderabad, Kandi, Sangareddy, Telangana 502284, India
| | - Eerappa Rajakumara
- Macromolecular Structural Biology Lab, Department of Biotechnology, Indian Institute of Technology Hyderabad, Kandi, Sangareddy, Telangana 502284, India.
| |
Collapse
|
2
|
Zhu F, Xu D. Predicting gene signature in breast cancer patients with multiple machine learning models. Discov Oncol 2024; 15:516. [PMID: 39352418 PMCID: PMC11445210 DOI: 10.1007/s12672-024-01386-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 09/23/2024] [Indexed: 10/04/2024] Open
Abstract
AIMS The aim of this study was to predict gene signatures in breast cancer patients using multiple machine learning models. METHODS In this study, we first collated and merged the datasets GSE54002 and GSE22820, obtaining a gene expression matrix comprising 16,820 genes (including 593 breast cancer (BC) samples and 26 normal control (NC) samples). Subsequently, we performed enrichment analyses using Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG), and Disease Ontology (DO). RESULTS We identified 177 differentially expressed genes (DEGs), including 40 up-regulated and 137 down-regulated genes, through differential expression analysis. The GO enrichment results indicated that these genes are primarily involved in extracellular matrix organization, positive regulation of nervous system development, collagen-containing extracellular matrix, heparin binding, glycosaminoglycan binding, and Wnt protein binding, among others. KEGG enrichment analysis revealed that the DEGs were primarily associated with pathways such as focal adhesion, the PI3K-Akt signaling pathway, and human papillomavirus infection. DO enrichment analysis showed that the DEGs play a significant role in regulating diseases such as intestinal disorders, nephritis, and dermatitis. Further, through LASSO regression analysis and SVM-RFE algorithm analysis, we identified 9 key feature DEGs (CF-DEGs): ANGPTL7, TSHZ2, SDPR, CLCA4, PAMR1, MME, CXCL2, ADAMTS5, and KIT. Additionally, ROC curve analysis demonstrated that these CF-DEGs serve as a reliable diagnostic index. Finally, using the CIBERSORT algorithm, we analyzed the infiltration of immune cells and the associations between CF-DEGs and immune cell infiltration across all samples. CONCLUSIONS Our findings provide new insights into the molecular functions and metabolic pathways involved in breast cancer, potentially aiding in the discovery of new diagnostic and immunotherapeutic biomarkers.
Collapse
Affiliation(s)
- Fangfang Zhu
- First Affiliated Hospital of Huzhou University, No.158, Guangchang Hou Road, Huzhou, 313000, Zhejiang, People's Republic of China
| | - Dafang Xu
- First Affiliated Hospital of Huzhou University, No.158, Guangchang Hou Road, Huzhou, 313000, Zhejiang, People's Republic of China.
| |
Collapse
|
3
|
Adebayo AK, Bhat-Nakshatri P, Davis C, Angus SP, Erdogan C, Gao H, Green N, Kumar B, Liu Y, Nakshatri H. Oxygen tension-dependent variability in the cancer cell kinome impacts signaling pathways and response to targeted therapies. iScience 2024; 27:110068. [PMID: 38872973 PMCID: PMC11170190 DOI: 10.1016/j.isci.2024.110068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 04/05/2024] [Accepted: 05/17/2024] [Indexed: 06/15/2024] Open
Abstract
Most cells in solid tumors are exposed to oxygen levels between 0.5% and 5%. We developed an approach that allows collection, processing, and evaluation of cancer and non-cancer cells under physioxia, while preventing exposure to ambient air. This aided comparison of baseline and drug-induced changes in signaling pathways under physioxia and ambient oxygen. Using tumor cells from transgenic models of breast cancer and cells from breast tissues of clinically breast cancer-free women, we demonstrate oxygen-dependent differences in cell preference for epidermal growth factor receptor (EGFR) or platelet-derived growth factor receptor beta (PDGFRβ) signaling. Physioxia caused PDGFRβ-mediated activation of AKT and extracellular regulated kinase (ERK) that reduced sensitivity to EGFR and phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit alpha (PIK3CA) inhibition and maintained PDGFRβ+ epithelial-mesenchymal hybrid cells with potential cancer stem cell (CSC) properties. Cells in ambient air displayed differential EGFR activation and were more sensitive to targeted therapies. Our data emphasize the importance of oxygen considerations in preclinical cancer research to identify effective drug targets and develop combination therapy regimens.
Collapse
Affiliation(s)
- Adedeji K. Adebayo
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Indiana University Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | | | - Christopher Davis
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Steven P. Angus
- Indiana University Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Cihat Erdogan
- Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Hongyu Gao
- Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Nick Green
- Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Brijesh Kumar
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Yunlong Liu
- Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Harikrishna Nakshatri
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Indiana University Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Roudebush VA Medical Center, Indianapolis, IN 46202, USA
| |
Collapse
|
4
|
Fang Y, Zhang Q, Chen Z, Guo C, Wu J. Clinical significance and immune characteristics analysis of miR-221-3p and its key target genes related to epithelial-mesenchymal transition in breast cancer. Aging (Albany NY) 2024; 16:322-347. [PMID: 38189813 PMCID: PMC10817385 DOI: 10.18632/aging.205370] [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: 07/20/2023] [Accepted: 11/20/2023] [Indexed: 01/09/2024]
Abstract
BACKGROUND MicroRNA-221-3p (miR-221-3p) facilitates the advancement of breast cancer (BC) through the induction of epithelial-mesenchymal transition (EMT). Our research aimed to utilize bioinformatics to discover possible EMT-related target genes (ETGs) of miR-221-3p and examine their roles in breast cancer. METHODS We employed bioinformatics techniques to identify ten key ETGs of miR-221-3p. Subsequently, we conducted an extensive analysis of both miR-221-3p and the ten ETGs, including clinical significance and immune characteristics. RESULTS The expression of miR-221-3p was notably higher in Basal-like BC compared to other subtypes and adjacent normal tissue. Our pathway analysis suggested that miR-221-3p might regulate EMT through the MAPK signaling pathway by targeting its ETGs. Among the ETGs, seven core genes (EGFR, IGF1, KDR, FGF2, KIT, FGFR1, and FGF1) exhibited downregulation in BC. Conversely, ERBB2, SDC1, and MMP14 showed upregulation in BC and displayed potential diagnostic value. The analysis of prognostication indicated that increased levels of SDC1 and MMP14 were correlated with an unfavorable prognosis, whereas elevated expression of KIT was associated with a more favorable prognosis. The infiltration of various immune cells and the expression of immune checkpoint genes (ICGs) exhibited positive correlations with most ETGs and miR-221-3p. SDC1 exhibited a greater tumor mutational burden (TMB) score, while ERBB2, KDR, FGF2, KIT, FGFR1, and FGF1 showed lower TMB scores. Furthermore, decreased ERBB2 and KDR expression levels were correlated with elevated microsatellite instability (MSI) scores. Elevated expression of ETGs was linked to decreased mRNA stemness indices (mRNAsi), whereas miR-221-3p displayed the opposite pattern. Most ETGs and miR-221-3p expression exhibited a negative correlation with IC50 values for drugs. Among the ETGs, amplification was the most significant genetic alteration, except for IGF1. CONCLUSION In conclusion, miR-221-3p acts as a unique indicator for Basal-like BC. The examination revealed ten essential ETGs of miR-221-3p, some of which show potential as diagnostic and prognostic markers. The in-depth examination of these ten ETGs and miR-221-3p indicates their participation in the development of BC, emphasizing their promise as innovative targets for therapy in BC patients.
Collapse
Affiliation(s)
- Yutong Fang
- The Breast Center, Cancer Hospital of Shantou University Medical College, Shantou 515041, Guangdong, China
- Department of Central Laboratory, Cancer Hospital of Shantou University Medical College, Shantou 515041, Guangdong, China
| | - Qunchen Zhang
- Department of Breast, Jiangmen Central Hospital, Jiangmen 529000, Guangdong, China
| | - Zexiao Chen
- The Breast Center, Cancer Hospital of Shantou University Medical College, Shantou 515041, Guangdong, China
- Department of Central Laboratory, Cancer Hospital of Shantou University Medical College, Shantou 515041, Guangdong, China
| | - Cuiping Guo
- The Breast Center, Cancer Hospital of Shantou University Medical College, Shantou 515041, Guangdong, China
- Department of Central Laboratory, Cancer Hospital of Shantou University Medical College, Shantou 515041, Guangdong, China
| | - Jundong Wu
- The Breast Center, Cancer Hospital of Shantou University Medical College, Shantou 515041, Guangdong, China
- Department of Central Laboratory, Cancer Hospital of Shantou University Medical College, Shantou 515041, Guangdong, China
| |
Collapse
|
5
|
Abdellateif MS, Bayoumi AK, Mohammed MA. c-Kit Receptors as a Therapeutic Target in Cancer: Current Insights. Onco Targets Ther 2023; 16:785-799. [PMID: 37790582 PMCID: PMC10544070 DOI: 10.2147/ott.s404648] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Accepted: 09/19/2023] [Indexed: 10/05/2023] Open
Abstract
c-Kit is a type III receptor tyrosine kinase (RTK) that has an essential role in various biological functions including gametogenesis, melanogenesis, hematopoiesis, cell survival, and apoptosis. c-KIT aberrations, either overexpression or loss-of-function mutations, have been implicated in the pathogenesis and development of many cancers, including gastrointestinal stromal tumors, mastocytosis, acute myeloid leukemia, breast, thyroid, and colorectal cancer, making c-KIT an attractive molecular target for the treatment of cancers. Therefore, a lot of effort has been put into investigating the utility of tyrosine kinase inhibitors for the management of c-KIT mutated tumors. This review of the literature illustrates the role of c-KIT mutations in many cancers, aiming to provide insights into the role of TKIs as a therapeutic option for cancer patients with c-KIT aberrations. In conclusion, c-KIT is implicated in different types of cancer, and it could be a successful molecular target; however, proper detection of the underlying mutation type is required before starting the appropriate personalized therapy.
Collapse
Affiliation(s)
- Mona S Abdellateif
- Medical Biochemistry and Molecular Biology, Cancer Biology Department, National Cancer Institute, Cairo University, Cairo, 11796, Egypt
| | - Ahmed K Bayoumi
- Paediatric Oncology Department, National Cancer Institute, Cairo University, Cairo, 11796, Egypt
- Children’s Cancer Hospital 57357, Cairo, 11617, Egypt
| | - Mohammed Aly Mohammed
- Medical Biochemistry and Molecular Biology, Cancer Biology Department, National Cancer Institute, Cairo University, Cairo, 11796, Egypt
| |
Collapse
|
6
|
D'Amico F, Graziano R, D'Aria F, Russomanno P, Di Fonzo S, Amato J, Pagano B. Cytosine epigenetic modifications and conformational changes in G-quadruplex DNA: An ultraviolet resonance Raman spectroscopy study. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2023; 300:122901. [PMID: 37244027 DOI: 10.1016/j.saa.2023.122901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 04/26/2023] [Accepted: 05/17/2023] [Indexed: 05/29/2023]
Abstract
Epigenetic modifications of DNA are known to play important regulatory roles in biological systems, especially in regulation of gene expression, and are associated with many types of human diseases, including cancer. Alternative DNA secondary structures, such as G-quadruplexes, can also influence gene transcription, thus suggesting that such structures may represent a distinctive layer of epigenetic information. G-quadruplex structures and DNA epigenetic modifications often go side by side, and recent evidence reveals that cytosine modifications within loops of G-quadruplexes can play a role in modulating their stability and structural polymorphism. Therefore, the development and validation of experimental techniques that can easily and reliably analyse G-quadruplex structures are highly desirable. In the present study, we propose to exploit the advantages of UV resonance Raman (UVRR) spectroscopy to investigate cytosine epigenetic modifications along with conformational changes in G-quadruplex-forming DNA. Our findings show that clear and specific spectral changes occur when there is a change in a G-quadruplex structure. Moreover, UVRR spectral analysis can indirectly distinguish the spectral variations occurring because of modifications in the guanine glycosidic conformations, as well as detect changes in the loops induced by H-bond formation or hydration of nitrogenous bases. The results further underscore the utility of UVRR spectroscopy for G-quadruplex structure elucidation under biologically relevant solution conditions.
Collapse
Affiliation(s)
- Francesco D'Amico
- Elettra-Sincrotrone Trieste S. C. p. A., Science Park, Trieste I-34149, Italy
| | - Raffaele Graziano
- Department of Pharmacy, University of Naples Federico II, Via D. Montesano 49, 80131 Naples, Italy
| | - Federica D'Aria
- Department of Pharmacy, University of Naples Federico II, Via D. Montesano 49, 80131 Naples, Italy
| | - Pasquale Russomanno
- Department of Pharmacy, University of Naples Federico II, Via D. Montesano 49, 80131 Naples, Italy
| | - Silvia Di Fonzo
- Elettra-Sincrotrone Trieste S. C. p. A., Science Park, Trieste I-34149, Italy
| | - Jussara Amato
- Department of Pharmacy, University of Naples Federico II, Via D. Montesano 49, 80131 Naples, Italy.
| | - Bruno Pagano
- Department of Pharmacy, University of Naples Federico II, Via D. Montesano 49, 80131 Naples, Italy.
| |
Collapse
|
7
|
Bhat SM, Prasad PR, Joshi MB. Novel insights into DNA methylation-based epigenetic regulation of breast tumor angiogenesis. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2023; 380:63-96. [PMID: 37657860 DOI: 10.1016/bs.ircmb.2023.04.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/03/2023]
Abstract
Breast tumors are highly vascularized and dependent on angiogenesis for growth, progression and metastasis. Like other solid tumors, vasculature in breast tumors also display leaky and tortuous phenotype and hence inhibit immune cell infiltration, show reduced efficacy to anticancer drugs and radiotherapy. Epigenetic reprogramming including significant alterations in DNA methylation in tumor and stromal cells generate an imbalance in expression of pro- and anti-angiogenic factors and subsequently lead to disordered angiogenesis. Hence, understanding DNA methylation-based regulation of angiogenesis in breast tumors may open new avenues for designing therapeutic targets. Our present review manuscript summarized contemporary knowledge of influence of DNA methylation in regulating angiogenesis. Further, we identified novel set of pro-angiogenic genes enriched in endothelial cells which are coregulated with DNMT isoforms in breast tumors and harboring CpG islands. Our analysis revealed promoters of pro-angiogenic genes were hypomethylated and anti-angiogenic genes were hypermethylated in tumors and further reflected on their expression patterns. Interestingly, promoter DNA methylation intensities of novel set of pro-angiogenic genes significantly correlated to patient survival outcome.
Collapse
Affiliation(s)
- Sharath Mohan Bhat
- Department of Ageing Research, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, India
| | - Palla Ranga Prasad
- Department of Ageing Research, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, India
| | - Manjunath B Joshi
- Department of Ageing Research, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, India.
| |
Collapse
|
8
|
Tóthová Z, Šemeláková M, Bhide K, Bhide M, Kováč A, Majerová P, Kvaková M, Štofilová J, Solárová Z, Solár P. Differentially Expressed Genes Induced by Erythropoietin Receptor Overexpression in Rat Mammary Adenocarcinoma RAMA 37-28 Cells. Int J Mol Sci 2023; 24:ijms24108482. [PMID: 37239828 DOI: 10.3390/ijms24108482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 04/25/2023] [Accepted: 05/02/2023] [Indexed: 05/28/2023] Open
Abstract
The erythropoietin receptor (EPOR) is a transmembrane type I receptor with an essential role in the proliferation and differentiation of erythroid progenitors. Besides its function during erythropoiesis, EPOR is expressed and has protective effect in various non-hematopoietic tissues, including tumors. Currently, the advantageous aspect of EPOR related to different cellular events is still under scientific investigation. Besides its well-known effect on cell proliferation, apoptosis and differentiation, our integrative functional study revealed its possible associations with metabolic processes, transport of small molecules, signal transduction and tumorigenesis. Comparative transcriptome analysis (RNA-seq) identified 233 differentially expressed genes (DEGs) in EPOR overexpressed RAMA 37-28 cells compared to parental RAMA 37 cells, whereas 145 genes were downregulated and 88 upregulated. Of these, for example, GPC4, RAP2C, STK26, ZFP955A, KIT, GAS6, PTPRF and CXCR4 were downregulated and CDH13, NR0B1, OCM2, GPM6B, TM7SF3, PARVB, VEGFD and STAT5A were upregulated. Surprisingly, two ephrin receptors, EPHA4 and EPHB3, and EFNB1 ligand were found to be upregulated as well. Our study is the first demonstrating robust differentially expressed genes evoked by simple EPOR overexpression without the addition of erythropoietin ligand in a manner which remains to be elucidated.
Collapse
Affiliation(s)
- Zuzana Tóthová
- Department of Medical Biology, Faculty of Medicine, P.J. Šafárik University in Košice, 04001 Košice, Slovakia
| | - Martina Šemeláková
- Department of Medical Biology, Faculty of Medicine, P.J. Šafárik University in Košice, 04001 Košice, Slovakia
| | - Katarína Bhide
- Laboratory of Biomedical Microbiology and Immunology, University of Veterinary Medicine and Pharmacy in Košice, 04001 Košice, Slovakia
| | - Mangesh Bhide
- Laboratory of Biomedical Microbiology and Immunology, University of Veterinary Medicine and Pharmacy in Košice, 04001 Košice, Slovakia
- Institute of Neuroimmunology, Slovak Academy of Sciences, 84510 Bratislava, Slovakia
| | - Andrej Kováč
- Institute of Neuroimmunology, Slovak Academy of Sciences, 84510 Bratislava, Slovakia
| | - Petra Majerová
- Institute of Neuroimmunology, Slovak Academy of Sciences, 84510 Bratislava, Slovakia
| | - Monika Kvaková
- Department of Experimental Medicine, Faculty of Medicine, P.J. Šafárik University in Košice, 04001 Košice, Slovakia
| | - Jana Štofilová
- Department of Experimental Medicine, Faculty of Medicine, P.J. Šafárik University in Košice, 04001 Košice, Slovakia
| | - Zuzana Solárová
- Department of Pharmacology, Faculty of Medicine, P.J. Šafárik University in Košice, 04001 Košice, Slovakia
| | - Peter Solár
- Department of Medical Biology, Faculty of Medicine, P.J. Šafárik University in Košice, 04001 Košice, Slovakia
| |
Collapse
|
9
|
Todhunter ME, Miyano M, Carlson EG, Hinz S, LaBarge MA. Sustained postconfluent culture of human mammary epithelial cells enriches for luminal and c-Kit+ subtypes. Breast Cancer Res 2023; 25:6. [PMID: 36653787 PMCID: PMC9847146 DOI: 10.1186/s13058-022-01595-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 12/16/2022] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND A challenge in human mammary epithelial cell (HMEC) culture is sustaining the representation of competing luminal, myoepithelial, and progenitor lineages over time. As cells replicate in culture, myoepithelial cells come to dominate the composition of the culture with serial passaging. This drift in composition presents a challenge for studying luminal and progenitor cells, which are prospective cells of origin for most breast cancer subtypes. METHODS We demonstrate the use of postconfluent culture on HMECs. Postconfluent culture entails culturing HMECs for 2-5 weeks without passaging but maintaining frequent feedings in low-stress M87A culture medium. In contrast, standard HMEC culture entails enzymatic subculturing every 3-5 days to maintain subconfluent density. RESULTS When compared to standard HMEC culture, postconfluent culture yields increased proportions of luminal cells and c-Kit+ progenitor cells. Postconfluent cultures develop a distinct multilayered morphology with individual cells showing decreased physical deformability as compared to cells in standard culture. Gene expression analysis of postconfluent cells shows increased expression of lineage-specific markers and extracellular matrix components. CONCLUSIONS Postconfluent culture is a novel, useful strategy for altering the lineage composition of HMECs, by increasing the proportional representation of luminal and progenitor cells. We speculate that postconfluent culture creates a microenvironment with cellular composition closer to the physiological state and eases the isolation of scarce cell subtypes. As such, postconfluent culture is a valuable tool for researchers using HMECs for breast cancer research.
Collapse
Affiliation(s)
- Michael E. Todhunter
- grid.410425.60000 0004 0421 8357Department of Population Sciences, Beckman Research Institute at City of Hope, 1500 E. Duarte Rd, Duarte, CA 91010 USA
| | - Masaru Miyano
- grid.410425.60000 0004 0421 8357Department of Population Sciences, Beckman Research Institute at City of Hope, 1500 E. Duarte Rd, Duarte, CA 91010 USA
| | - Eric G. Carlson
- grid.410425.60000 0004 0421 8357Department of Population Sciences, Beckman Research Institute at City of Hope, 1500 E. Duarte Rd, Duarte, CA 91010 USA ,grid.410425.60000 0004 0421 8357Irell and Manella Graduate School of Biological Sciences, City of Hope, 1500 E. Duarte Rd, Duarte, CA 91010 USA
| | - Stefan Hinz
- grid.410425.60000 0004 0421 8357Department of Population Sciences, Beckman Research Institute at City of Hope, 1500 E. Duarte Rd, Duarte, CA 91010 USA
| | - Mark A. LaBarge
- grid.410425.60000 0004 0421 8357Department of Population Sciences, Beckman Research Institute at City of Hope, 1500 E. Duarte Rd, Duarte, CA 91010 USA
| |
Collapse
|
10
|
Klein-Rodewald T, Micklich K, Sanz-Moreno A, Tost M, Calzada-Wack J, Adler T, Klaften M, Sabrautzki S, Aigner B, Kraiger M, Gailus-Durner V, Fuchs H, Gründer A, Pahl H, Wolf E, Hrabe de Angelis M, Rathkolb B, Rozman J, Puk O, Schrewe A, Schulz H, Adamski J, Busch DH, Esposito I, Wurst W, Stoeger C, Gründer A, Pahl H, Wolf E, Hrabe de Angelis M, Rathkolb B. New C3H Kit N824K/WT cancer mouse model develops late-onset malignant mammary tumors with high penetrance. Sci Rep 2022; 12:19793. [PMID: 36396684 PMCID: PMC9671887 DOI: 10.1038/s41598-022-23218-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 10/26/2022] [Indexed: 11/18/2022] Open
Abstract
Gastro-intestinal stromal tumors and acute myeloid leukemia induced by activating stem cell factor receptor tyrosine kinase (KIT) mutations are highly malignant. Less clear is the role of KIT mutations in the context of breast cancer. Treatment success of KIT-induced cancers is still unsatisfactory because of primary or secondary resistance to therapy. Mouse models offer essential platforms for studies on molecular disease mechanisms in basic cancer research. In the course of the Munich N-ethyl-N-nitrosourea (ENU) mutagenesis program a mouse line with inherited polycythemia was established. It carries a base-pair exchange in the Kit gene leading to an amino acid exchange at position 824 in the activation loop of KIT. This KIT variant corresponds to the N822K mutation found in human cancers, which is associated with imatinib-resistance. C3H KitN824K/WT mice develop hyperplasia of interstitial cells of Cajal and retention of ingesta in the cecum. In contrast to previous Kit-mutant models, we observe a benign course of gastrointestinal pathology associated with prolonged survival. Female mutants develop mammary carcinomas at late onset and subsequent lung metastasis. The disease model complements existing oncology research platforms. It allows for addressing the role of KIT mutations in breast cancer and identifying genetic and environmental modifiers of disease progression.
Collapse
Affiliation(s)
- Tanja Klein-Rodewald
- grid.4567.00000 0004 0483 2525Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Kateryna Micklich
- grid.4567.00000 0004 0483 2525Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Adrián Sanz-Moreno
- grid.4567.00000 0004 0483 2525Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Monica Tost
- grid.4567.00000 0004 0483 2525Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Julia Calzada-Wack
- grid.4567.00000 0004 0483 2525Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Thure Adler
- grid.4567.00000 0004 0483 2525Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Matthias Klaften
- grid.4567.00000 0004 0483 2525Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany ,Present Address: amcure GmbH, Herrman-von-Helmholtz-Platz 1, 76344 Eggenstein-Leopoldshafen, Germany
| | - Sibylle Sabrautzki
- grid.4567.00000 0004 0483 2525Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany ,grid.4567.00000 0004 0483 2525Research Unit Comparative Medicine, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Bernhard Aigner
- grid.5252.00000 0004 1936 973XInstitute of Molecular Animal Breeding and Biotechnology, Gene Center, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Markus Kraiger
- grid.4567.00000 0004 0483 2525Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Valerie Gailus-Durner
- grid.4567.00000 0004 0483 2525Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Helmut Fuchs
- grid.4567.00000 0004 0483 2525Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | | | - Albert Gründer
- grid.7708.80000 0000 9428 7911Section of Molecular Hematology, Department of Hematology/Oncology, Universitäts Klinikum Freiburg, Freiburg, Germany
| | - Heike Pahl
- grid.7708.80000 0000 9428 7911Section of Molecular Hematology, Department of Hematology/Oncology, Universitäts Klinikum Freiburg, Freiburg, Germany
| | - Eckhard Wolf
- grid.5252.00000 0004 1936 973XInstitute of Molecular Animal Breeding and Biotechnology, Gene Center, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Martin Hrabe de Angelis
- grid.4567.00000 0004 0483 2525Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany ,grid.452622.5German Center for Diabetes Research (DZD), Neuherberg, Germany ,grid.6936.a0000000123222966Chair of Experimental Genetics, TUM School of Life Sciences, Technische Universität München, Freising, Germany
| | - Birgit Rathkolb
- grid.4567.00000 0004 0483 2525Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany ,grid.5252.00000 0004 1936 973XInstitute of Molecular Animal Breeding and Biotechnology, Gene Center, Ludwig-Maximilians-Universität München, Munich, Germany ,grid.452622.5German Center for Diabetes Research (DZD), Neuherberg, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
11
|
c-Kit Induces Migration of Triple-Negative Breast Cancer Cells and Is a Promising Target for Tyrosine Kinase Inhibitor Treatment. Int J Mol Sci 2022; 23:ijms23158702. [PMID: 35955836 PMCID: PMC9369219 DOI: 10.3390/ijms23158702] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 07/14/2022] [Accepted: 07/16/2022] [Indexed: 11/24/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is associated with a poor prognosis and the absence of targeted therapy. c-Kit, a receptor tyrosine kinase (RTK), is considered a molecular target for anticancer drugs. Tyrosine kinase inhibitors (TKIs) recognizing c-Kit are used for the treatment of c-Kit-expressing tumors. However, the expression, function, and therapeutic potential of c-Kit have been little explored in TNBC. Here, we studied the expression and effects of c-Kit in TNBC through in vitro and in silico analysis, and evaluated the response to TKIs targeting c-Kit. Analysis of TNBC cells showed the expression of functional c-Kit at the cell membrane. The stimulation of c-Kit with its ligand induced the activation of STAT3, Akt, and ERK1/2, increasing cell migration, but had no effect on cell proliferation or response to Doxorubicin. Analysis of public datasets showed that the expression of c-Kit in tumors was not associated with patient survival. Finally, TNBC cells were susceptible to TKIs, in particular the effect of Nilotinib was stronger than Doxorubicin in all cell lines. In conclusion, TNBC cells express functional c-Kit, which is a targetable molecule, and show a strong response to Nilotinib that may be considered a candidate drug for the treatment of TNBC.
Collapse
|
12
|
Albogami S. Comprehensive analysis of gene expression profiles to identify differential prognostic factors of primary and metastatic breast cancer. Saudi J Biol Sci 2022; 29:103318. [PMID: 35677896 PMCID: PMC9168623 DOI: 10.1016/j.sjbs.2022.103318] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 04/17/2022] [Accepted: 05/19/2022] [Indexed: 12/21/2022] Open
Abstract
Breast cancer accounts for nearly half of all cancer-related deaths in women worldwide. However, the molecular mechanisms that lead to tumour development and progression remain poorly understood and there is a need to identify candidate genes associated with primary and metastatic breast cancer progression and prognosis. In this study, candidate genes associated with prognosis of primary and metastatic breast cancer were explored through a novel bioinformatics approach. Primary and metastatic breast cancer tissues and adjacent normal breast tissues were evaluated to identify biomarkers characteristic of primary and metastatic breast cancer. The Cancer Genome Atlas-breast invasive carcinoma (TCGA-BRCA) dataset (ID: HS-01619) was downloaded using the mRNASeq platform. Genevestigator 8.3.2 was used to analyse TCGA-BRCA gene expression profiles between the sample groups and identify the differentially-expressed genes (DEGs) in each group. For each group, Gene Ontology and Kyoto Encyclopedia of Genes and Genomes pathway enrichment analyses were used to determine the function of DEGs. Networks of protein-protein interactions were constructed to identify the top hub genes with the highest degree of interaction. Additionally, the top hub genes were validated based on overall survival and immunohistochemistry using The Human Protein Atlas. Of the top 20 hub genes identified, four (KRT14, KIT, RAD51, and TTK) were considered as prognostic risk factors based on overall survival. KRT14 and KIT expression levels were upregulated while those of RAD51 and TTK were downregulated in patients with breast cancer. The four proposed candidate hub genes might aid in further understanding the molecular changes that distinguish primary breast tumours from metastatic tumours as well as help in developing novel therapeutics. Furthermore, they may serve as effective prognostic risk markers based on the strong correlation between their expression and patient overall survival.
Collapse
Key Words
- BC, breast cancer
- BP, biological process
- Breast cancer
- CC, cellular component
- CI, confidence interval
- DEG, differentially expressed gene
- Differentially expressed genes
- FDR, false discovery rate
- GEPIA, gene expression profiling interactive analysis
- GO, gene ontology
- HR, hazard ratio
- IDC, infiltrating ductal carcinoma
- KEGG, Kyoto Encyclopedia of Genes and Genomes
- MCODE, molecular complex detection
- MF, molecular function
- Metastasis
- OS, overall survival
- Overall survival
- PPI, protein-protein interaction
- Prognostic marker
- Protein-protein interaction
- RNA-Seq, RNA sequencing
- STRING, search tool for the retrieval of interacting genes
- TCGA-BRCA, The Cancer Genome Atlas-breast invasive carcinoma
Collapse
Affiliation(s)
- Sarah Albogami
- Department of Biotechnology, College of Science, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
| |
Collapse
|
13
|
Vignjević Petrinović S, Jauković A, Milošević M, Bugarski D, Budeč M. Targeting Stress Erythropoiesis Pathways in Cancer. Front Physiol 2022; 13:844042. [PMID: 35694408 PMCID: PMC9174937 DOI: 10.3389/fphys.2022.844042] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Accepted: 05/09/2022] [Indexed: 11/13/2022] Open
Abstract
Cancer-related anemia (CRA) is a common multifactorial disorder that adversely affects the quality of life and overall prognosis in patients with cancer. Safety concerns associated with the most common CRA treatment options, including intravenous iron therapy and erythropoietic-stimulating agents, have often resulted in no or suboptimal anemia management for many cancer patients. Chronic anemia creates a vital need to restore normal erythropoietic output and therefore activates the mechanisms of stress erythropoiesis (SE). A growing body of evidence demonstrates that bone morphogenetic protein 4 (BMP4) signaling, along with glucocorticoids, erythropoietin, stem cell factor, growth differentiation factor 15 (GDF15) and hypoxia-inducible factors, plays a pivotal role in SE. Nevertheless, a chronic state of SE may lead to ineffective erythropoiesis, characterized by the expansion of erythroid progenitor pool, that largely fails to differentiate and give rise to mature red blood cells, further aggravating CRA. In this review, we summarize the current state of knowledge on the emerging roles for stress erythroid progenitors and activated SE pathways in tumor progression, highlighting the urgent need to suppress ineffective erythropoiesis in cancer patients and develop an optimal treatment strategy as well as a personalized approach to CRA management.
Collapse
Affiliation(s)
- Sanja Vignjević Petrinović
- Laboratory for Neuroendocrinology, Institute for Medical Research, National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Aleksandra Jauković
- Laboratory for Experimental Hematology and Stem Cells, Institute for Medical Research, National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Maja Milošević
- Laboratory for Neuroendocrinology, Institute for Medical Research, National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Diana Bugarski
- Laboratory for Experimental Hematology and Stem Cells, Institute for Medical Research, National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Mirela Budeč
- Laboratory for Neuroendocrinology, Institute for Medical Research, National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
14
|
Cao YN, Li QZ, Liu YX. Discovered Key CpG Sites by Analyzing DNA Methylation and Gene Expression in Breast Cancer Samples. Front Cell Dev Biol 2022; 10:815843. [PMID: 35178391 PMCID: PMC8844453 DOI: 10.3389/fcell.2022.815843] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 01/06/2022] [Indexed: 12/24/2022] Open
Abstract
Breast cancer is the most common cancer in the world, and DNA methylation plays a key role in the occurrence and development of breast cancer. However, the effect of DNA methylation in different gene functional regions on gene expression and the effect of gene expression on breast cancer is not completely clear. In our study, we computed and analyzed DNA methylation, gene expression, and clinical data in the TCGA database. Firstly, we calculated the distribution of abnormal DNA methylated probes in 12 regions, found the abnormal DNA methylated probes in down-regulated genes were highly enriched, and the number of hypermethylated probes in the promoter region was 6.5 times than that of hypomethylated probes. Secondly, the correlation coefficients between abnormal DNA methylated values in each functional region of differentially expressed genes and gene expression values were calculated. Then, co-expression analysis of differentially expressed genes was performed, 34 hub genes in cancer-related pathways were obtained, of which 11 genes were regulated by abnormal DNA methylation. Finally, a multivariate Cox regression analysis was performed on 27 probes of 11 genes. Three DNA methylation probes (cg13569051 and cg14399183 of GSN, and cg25274503 of CAV2) related to survival were used to construct a prognostic model, which has a good prognostic ability. Furthermore, we found that the cg25274503 hypermethylation in the promoter region inhibited the expression of the CAV2, and the hypermethylation of cg13569051 and cg14399183 in the 5′UTR region inhibited the expression of GSN. These results may provide possible molecular targets for breast cancer.
Collapse
Affiliation(s)
- Yan-Ni Cao
- Laboratory of Theoretical Biophysics, School of Physical Science and Technology, Inner Mongolia University, Hohhot, China
| | - Qian-Zhong Li
- Laboratory of Theoretical Biophysics, School of Physical Science and Technology, Inner Mongolia University, Hohhot, China.,The State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Inner Mongolia University, Hohhot, China
| | - Yu-Xian Liu
- Laboratory of Theoretical Biophysics, School of Physical Science and Technology, Inner Mongolia University, Hohhot, China
| |
Collapse
|
15
|
Ercoskun P, Yuce Kahraman C, Ozkan G, Tatar A. Genetic Characterization of Hereditary Cancer Syndromes Based on Targeted Next-Generation Sequencing. Mol Syndromol 2021; 13:123-131. [DOI: 10.1159/000518927] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 08/06/2021] [Indexed: 11/19/2022] Open
Abstract
A hereditary cancer syndrome is a genetic predisposition to cancer caused by a germline mutation in cancer-related genes. Identifying the disease-causing variant is important for both the patient and relatives at risk in cancer families because this could be a guide in treatment and secondary cancer prevention. In this study, hereditary cancer panel harboring cancer-related genes was performed on MiSeq Illumina NGS system from peripheral blood samples. Sequencing files were fed into a cloud-based data analysis pipeline. Reportable variants were classified according to the American College of Medical Genetics and Genomics guidelines. Three hundred five individuals were included in the study. Different pathogenic/likely pathogenic variants were detected in 75 individuals. The majority of these variants were in the <i>MUTYH</i>, <i>BRCA2</i>, and <i>CHEK2</i> genes. Nine novel pathogenic/likely pathogenic variants were identified in <i>BRCA1</i>, <i>BRCA2</i>, <i>GALNT12</i>, <i>ATM</i>, <i>MLH1</i>, <i>MSH2</i>, <i>APC</i>, and <i>KIT</i> genes. We obtained interesting and novel variants which could be related to hereditary cancer, and this study confirmed that NGS is an indispensable method for the risk assessment in cancer families.
Collapse
|
16
|
KIT Expression Is Regulated by DNA Methylation in Uveal Melanoma Tumors. Int J Mol Sci 2021; 22:ijms221910748. [PMID: 34639089 PMCID: PMC8509522 DOI: 10.3390/ijms221910748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 09/27/2021] [Accepted: 09/30/2021] [Indexed: 11/21/2022] Open
Abstract
Uveal melanoma (UM) is an ocular tumor with a dismal prognosis. Despite the availability of precise molecular and cytogenetic techniques, clinicopathologic features with limited accuracy are widely used to predict metastatic potential. In 51 UM tissues, we assessed a correlation between the expression of nine proteins evaluated by immunohistochemistry (IHC) (Melan-A, S100, HMB45, Cyclin D1, Ki-67, p53, KIT, BCL2, and AIFM1) and the presence of UM-specific chromosomal rearrangements measured by multiplex ligation-dependent probe amplification (MLPA), to find IHC markers with increased prognostic information. Furthermore, mRNA expression and DNA methylation values were extracted from the whole-genome data, achieved by analyzing 22 fresh frozen UM tissues. KIT positivity was associated with monosomy 3, increasing the risk of poor prognosis more than 17-fold (95% CI 1.53–198.69, p = 0.021). A strong negative correlation was identified between mRNA expression and DNA methylation values for 12 of 20 analyzed positions, five located in regulatory regions of the KIT gene (r = −0.658, p = 0.001; r = −0.662, p = 0.001; r = −0.816; p < 0.001; r = −0.689, p = 0.001; r = −0.809, p < 0.001, respectively). DNA methylation β values were also inversely associated with KIT protein expression (p = 0.001; p = 0.001; p = 0.015; p = 0.025; p = 0.002). Our findings, showing epigenetic deregulation of KIT expression, may contribute to understanding the past failure to therapeutically target KIT in UM.
Collapse
|
17
|
Valdivia G, Alonso-Diez Á, Pérez-Alenza D, Peña L. From Conventional to Precision Therapy in Canine Mammary Cancer: A Comprehensive Review. Front Vet Sci 2021; 8:623800. [PMID: 33681329 PMCID: PMC7925635 DOI: 10.3389/fvets.2021.623800] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 01/11/2021] [Indexed: 12/16/2022] Open
Abstract
Canine mammary tumors (CMTs) are the most common neoplasm in intact female dogs. Canine mammary cancer (CMC) represents 50% of CMTs, and besides surgery, which is the elective treatment, additional targeted and non-targeted therapies could offer benefits in terms of survival to these patients. Also, CMC is considered a good spontaneous intermediate animal model for the research of human breast cancer (HBC), and therefore, the study of new treatments for CMC is a promising field in comparative oncology. Dogs with CMC have a comparable disease, an intact immune system, and a much shorter life span, which allows the achievement of results in a relatively short time. Besides conventional chemotherapy, innovative therapies have a large niche of opportunities. In this article, a comprehensive review of the current research in adjuvant therapies for CMC is conducted to gather available information and evaluate the perspectives. Firstly, updates are provided on the clinical-pathological approach and the use of conventional therapies, to delve later into precision therapies against therapeutic targets such as hormone receptors, tyrosine kinase receptors, p53 tumor suppressor gene, cyclooxygenases, the signaling pathways involved in epithelial-mesenchymal transition, and immunotherapy in different approaches. A comparison of the different investigations on targeted therapies in HBC is also carried out. In the last years, the increasing number of basic research studies of new promising therapeutic agents on CMC cell lines and CMC mouse xenografts is outstanding. As the main conclusion of this review, the lack of effort to bring the in vitro studies into the field of applied clinical research emerges. There is a great need for well-planned large prospective randomized clinical trials in dogs with CMC to obtain valid results for both species, humans and dogs, on the use of new therapies. Following the One Health concept, human and veterinary oncology will have to join forces to take advantage of both the economic and technological resources that are invested in HBC research, together with the innumerable advantages of dogs with CMC as a spontaneous animal model.
Collapse
Affiliation(s)
- Guillermo Valdivia
- Department Animal Medicine, Surgery and Pathology, Veterinary School, Complutense University of Madrid, Madrid, Spain
- Mammary Oncology Unit, Complutense Veterinary Teaching Hospital, Complutense University of Madrid, Madrid, Spain
| | - Ángela Alonso-Diez
- Department Animal Medicine, Surgery and Pathology, Veterinary School, Complutense University of Madrid, Madrid, Spain
- Mammary Oncology Unit, Complutense Veterinary Teaching Hospital, Complutense University of Madrid, Madrid, Spain
| | - Dolores Pérez-Alenza
- Department Animal Medicine, Surgery and Pathology, Veterinary School, Complutense University of Madrid, Madrid, Spain
- Mammary Oncology Unit, Complutense Veterinary Teaching Hospital, Complutense University of Madrid, Madrid, Spain
| | - Laura Peña
- Department Animal Medicine, Surgery and Pathology, Veterinary School, Complutense University of Madrid, Madrid, Spain
- Mammary Oncology Unit, Complutense Veterinary Teaching Hospital, Complutense University of Madrid, Madrid, Spain
| |
Collapse
|
18
|
Ruksha TG, Sergeeva EY, Fefelova YA, Khorzhevsky VA. [The significance of C-KIT gene mutations in the diagnosis and prognosis of malignant tumors]. Arkh Patol 2021; 83:61-68. [PMID: 34278763 DOI: 10.17116/patol20218304161] [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] [Indexed: 06/13/2023]
Abstract
Mutations in the C-KIT gene encoding type III receptor tyrosine kinase that regulates cellular processes, such as differentiation, survival, proliferation, migration, and apoptosis, are found in some neoplasms: gastrointestinal stromal tumor, mastocytosis, melanoma, breast carcinomas, myeloid leukemias, and a number of others. Tumors that exhibit these mutations are sensitive to therapy with tyrosine kinase inhibitors, which makes it necessary to correctly identify the mutation status by C-KIT in order to apply a personalized approach to therapy. This literature review shows that the type and localization of the C-KIT gene mutation are of crucial prognostic value and significance in choosing drugs for antitumor therapy, but traditional diagnostic methods fail to determine accurate mutation characteristics. Routine sequencing techniques focus on identifying the gene mutations associated with specific cellular processes, such as DNA damage and repair. The emergence of next-generation sequencing techniques has solved this problem, making it possible to fully analyze the genome of a malignant neoplasm, with constant screening for new mutations that appear as the tumor develops, affect the prognosis of the disease, and change its sensitivity to the antitumor therapy.
Collapse
Affiliation(s)
- T G Ruksha
- Prof. V.F. Voino-Yasenetsky Krasnoyarsk State Medical University of the Ministry of Health of Russia, Krasnoyarsk, Russia
| | - E Yu Sergeeva
- Prof. V.F. Voino-Yasenetsky Krasnoyarsk State Medical University of the Ministry of Health of Russia, Krasnoyarsk, Russia
| | - Yu A Fefelova
- Prof. V.F. Voino-Yasenetsky Krasnoyarsk State Medical University of the Ministry of Health of Russia, Krasnoyarsk, Russia
| | - V A Khorzhevsky
- Prof. V.F. Voino-Yasenetsky Krasnoyarsk State Medical University of the Ministry of Health of Russia, Krasnoyarsk, Russia
| |
Collapse
|
19
|
de Gannes M, Ko CI, Zhang X, Biesiada J, Niu L, Koch SE, Medvedovic M, Rubinstein J, Puga A. Dioxin Disrupts Dynamic DNA Methylation Patterns in Genes That Govern Cardiomyocyte Maturation. Toxicol Sci 2020; 178:325-337. [PMID: 33017471 DOI: 10.1093/toxsci/kfaa153] [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] [Indexed: 12/21/2022] Open
Abstract
Congenital heart disease (CHD), the leading birth defect worldwide, has a largely unknown etiology, likely to result from complex interactions between genetic and environmental factors during heart development, at a time when the heart adapts to diverse physiological and pathophysiological conditions. Crucial among these is the regulation of cardiomyocyte development and postnatal maturation, governed by dynamic changes in DNA methylation. Previous work from our laboratory has shown that exposure to the environmental toxicant tetrachlorodibenzo-p-dioxin (TCDD) disrupts several molecular networks responsible for heart development and function. To test the hypothesis that the disruption caused by TCDD in the heart results from changes in DNA methylation and gene expression patterns of cardiomyocytes, we established a stable mouse embryonic stem cell line expressing a puromycin resistance selectable marker under control of the cardiomyocyte-specific Nkx2-5 promoter. Differentiation of these cells in the presence of puromycin induces the expression of a large suite of cardiomyocyte-specific markers. To assess the consequences of TCDD treatment on gene expression and DNA methylation in these cardiomyocytes, we subjected them to transcriptome and methylome analyses in the presence of TCDD. Unlike control cardiomyocytes maintained in vehicle, the TCDD-treated cardiomyocytes showed extensive gene expression changes, with a significant correlation between differential RNA expression and DNA methylation in 111 genes, many of which are key elements of pathways that regulate cardiovascular development and function. Our findings provide an important clue toward the elucidation of the complex interactions between genetic and epigenetic mechanisms after developmental TCDD exposure that may contribute to CHD.
Collapse
Affiliation(s)
- Matthew de Gannes
- Department of Environmental Health and Center for Environmental Genetics
| | - Chia-I Ko
- Department of Environmental Health and Center for Environmental Genetics
| | - Xiang Zhang
- Department of Environmental Health and Center for Environmental Genetics
| | - Jacek Biesiada
- Department of Environmental Health and Center for Environmental Genetics
| | - Liang Niu
- Department of Environmental Health and Center for Environmental Genetics
| | - Sheryl E Koch
- Department of Internal Medicine, Division of Cardiovascular Health and Disease, University of Cincinnati College of Medicine, Cincinnati, Ohio 45267
| | - Mario Medvedovic
- Department of Environmental Health and Center for Environmental Genetics
| | - Jack Rubinstein
- Department of Internal Medicine, Division of Cardiovascular Health and Disease, University of Cincinnati College of Medicine, Cincinnati, Ohio 45267
| | - Alvaro Puga
- Department of Environmental Health and Center for Environmental Genetics
| |
Collapse
|
20
|
Amirkhani Namagerdi A, d'Angelo D, Ciani F, Iannuzzi CA, Napolitano F, Avallone L, De Laurentiis M, Giordano A. Triple-Negative Breast Cancer Comparison With Canine Mammary Tumors From Light Microscopy to Molecular Pathology. Front Oncol 2020; 10:563779. [PMID: 33282730 PMCID: PMC7689249 DOI: 10.3389/fonc.2020.563779] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Accepted: 10/19/2020] [Indexed: 12/12/2022] Open
Abstract
Many similar characteristics in human and dog cancers including, spontaneous development, clinical presentation, tumor heterogeneity, disease progression, and response to standard therapies have promoted the approval of this comparative model as an alternative to mice. Breast cancer represents the second most frequent neoplasm in humans after lung cancer. Triple-negative breast cancers (TNBC) constitute around 15% of all cases of breast cancer and do not express estrogen receptor (ER), progesterone receptor (PR), and do not overexpress human epidermal growth factor receptor 2 (HER2). As a result, they do not benefit from hormonal or trastuzumab-based therapy. Patients with TNBC have worse overall survival than patients with non-TNBC. Lehmann and collaborators described six different molecular subtypes of TNBC which further demonstrated its transcriptional heterogeneity. This six TNBC subtype classification has therapeutic implications. Breast cancer is the second most frequent neoplasm in sexually intact female dogs after skin cancer. Canine mammary tumors are a naturally occurring heterogeneous group of cancers that have several features in common with human breast cancer (HBC). These similarities include etiology, signaling pathway activation, and histological classification. Molecularly CMTs are more like TNBCs, and therefore dogs are powerful spontaneous models of cancer to test new therapeutic approaches, particularly for human TNBCs. More malignant tumors of the breast are more often ER and PR negative in both humans and dogs. Promising breast cancer biomarkers in both humans and canines are cancer-associated stroma (CAS), circulating tumor cells and tumor DNA (ctDNA), exosomes and miRNAs, and metabolites.
Collapse
Affiliation(s)
| | - Danila d'Angelo
- Department of Veterinary Medicine and Animal Production, University of Naples Federico II, Naples, Italy
| | - Francesca Ciani
- Department of Veterinary Medicine and Animal Production, University of Naples Federico II, Naples, Italy
| | | | - Francesco Napolitano
- Department of Veterinary Medicine and Animal Production, University of Naples Federico II, Naples, Italy.,CCEINGE, Biotecnologie Avanzate, Naples, Italy
| | - Luigi Avallone
- Department of Veterinary Medicine and Animal Production, University of Naples Federico II, Naples, Italy
| | - Michelino De Laurentiis
- Breast Oncology Division, Istituto Nazionale Tumori IRCCS Fondazione G. Pascale, Naples, Italy
| | - Antonio Giordano
- Center for Biotechnology, College of Science and Technology, Sbarro Institute for Cancer Research and Molecular Medicine, Temple University, Philadelphia, PA, United States.,Department of Medical Biotechnologies, University of Siena, Siena, Italy
| |
Collapse
|
21
|
Intermittent fasting from dawn to sunset for four consecutive weeks induces anticancer serum proteome response and improves metabolic syndrome. Sci Rep 2020; 10:18341. [PMID: 33110154 PMCID: PMC7592042 DOI: 10.1038/s41598-020-73767-w] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2020] [Accepted: 09/18/2020] [Indexed: 12/11/2022] Open
Abstract
Metabolic syndrome is characterized by central obesity, insulin resistance, elevated blood pressure, and dyslipidemia. Metabolic syndrome is a significant risk factor for several common cancers (e.g., liver, colorectal, breast, pancreas). Pharmacologic treatments used for the components of the metabolic syndrome appear to be insufficient to control cancer development in subjects with metabolic syndrome. Murine models showed that cancer has the slowest progression when there is no food consumption during the daily activity phase. Intermittent fasting from dawn to sunset is a form of fasting practiced during human activity hours. To test the anticancer effect of intermittent fasting from dawn to sunset in metabolic syndrome, we conducted a pilot study in 14 subjects with metabolic syndrome who fasted (no eating or drinking) from dawn to sunset for more than 14 h daily for four consecutive weeks. We collected serum samples before 4-week intermittent fasting, at the end of 4th week during 4-week intermittent fasting and 1 week after 4-week intermittent fasting. We performed serum proteomic analysis using nano ultra-high performance liquid chromatography-tandem mass spectrometry. We found a significant fold increase in the levels of several tumor suppressor and DNA repair gene protein products (GP)s at the end of 4th week during 4-week intermittent fasting (CALU, INTS6, KIT, CROCC, PIGR), and 1 week after 4-week intermittent fasting (CALU, CALR, IGFBP4, SEMA4B) compared with the levels before 4-week intermittent fasting. We also found a significant reduction in the levels of tumor promoter GPs at the end of 4th week during 4-week intermittent fasting (POLK, CD109, CAMP, NIFK, SRGN), and 1 week after 4-week intermittent fasting (CAMP, PLAC1) compared with the levels before 4-week intermittent fasting. Fasting from dawn to sunset for four weeks also induced an anti-diabetes proteome response by upregulating the key regulatory proteins of insulin signaling at the end of 4th week during 4-week intermittent fasting (VPS8, POLRMT, IGFBP-5) and 1 week after 4-week intermittent fasting (PRKCSH), and an anti-aging proteome response by upregulating H2B histone proteins 1 week after 4-week intermittent fasting. Subjects had a significant reduction in body mass index, waist circumference, and improvement in blood pressure that co-occurred with the anticancer, anti-diabetes, and anti-aging serum proteome response. These findings suggest that intermittent fasting from dawn to sunset actively modulates the respective genes and can be an adjunct treatment in metabolic syndrome. Further studies are needed to test the intermittent fasting from dawn to sunset in the prevention and treatment of metabolic syndrome-induced cancers.
Collapse
|
22
|
Dang L, Wang Y, Shi C, Liao M, Sun Z, Fang S. A Potential Tumor Suppressor Gene Named miR-508-5p Inhibited the Proliferation and Invasion of Human Melanoma Cells by Targeting KIT. Technol Cancer Res Treat 2020; 19:1533033820951801. [PMID: 33000689 PMCID: PMC7533916 DOI: 10.1177/1533033820951801] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Melanoma is the main death cause of human skin cancer. Increasing evidences demonstrate that microRNAs act as key roles in mediating tumor occurrence and progression. MiR-508-5p has proved to participate in the development of various types of human malignancies. However, the role of miR-508-5p in melanoma remained unclear. In in vitro study, miR-508-5p level in peripheral blood samples of patients with melanoma and human melanoma A375 cells was downregulated compared to that in normal peripheral blood samples or normal human epidermal melanocytes (MHEM). MiR-508-5p overexpression significantly inhibited the cell proliferation, migration and invasion in A375 cells, and thus inhibiting KIT expression at both gene and protein levels. Furthermore, western blot analysis showed miR-508-5p reduced cell proliferation by targeting KIT to modulate RAS/RAF/MEK/ERK pathway. Taken together, we speculated that miR-508-5p functioned as an important suppressor in human melanoma by targeting KIT, suggesting miR-508-5p might be a promising tumor suppressor gene for further target therapies from bench to clinic.
Collapse
Affiliation(s)
- Lin Dang
- Department of Dermatology, Shenzhen People's Hospital, The Second Clinical Medical College of Jinan University, The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen, Guangdong Province, China
| | - Yan Wang
- Department of Pathology, Shenzhen People's Hospital, The Second Clinical Medical College of Jinan University, The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen, Guangdong Province, China
| | - Cuiping Shi
- Department of Dermatology, Shenzhen People's Hospital, The Second Clinical Medical College of Jinan University, The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen, Guangdong Province, China
| | - Mengsi Liao
- Department of Dermatology, Shenzhen People's Hospital, The Second Clinical Medical College of Jinan University, The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen, Guangdong Province, China
| | - Zhaojun Sun
- Department of Dermatology, Shenzhen People's Hospital, The Second Clinical Medical College of Jinan University, The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen, Guangdong Province, China
| | - Sining Fang
- Department of Dermatology, Shenzhen People's Hospital, The Second Clinical Medical College of Jinan University, The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen, Guangdong Province, China
| |
Collapse
|
23
|
Pham DDM, Guhan S, Tsao H. KIT and Melanoma: Biological Insights and Clinical Implications. Yonsei Med J 2020; 61:562-571. [PMID: 32608199 PMCID: PMC7329741 DOI: 10.3349/ymj.2020.61.7.562] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 06/11/2020] [Indexed: 01/15/2023] Open
Abstract
Melanoma, originating from epidermal melanocytes, is a heterogeneous disease that has the highest mortality rate among all types of skin cancers. Numerous studies have revealed the cause of this cancer as related to various somatic driver mutations, including alterations in KIT-a proto-oncogene encoding for a transmembrane receptor tyrosine kinase. Although accounting for only 3% of all melanomas, mutations in c-KIT are mostly derived from acral, mucosal, and chronically sun-damaged melanomas. As an important factor for cell differentiation, proliferation, and survival, inhibition of c-KIT has been exploited for clinical trials in advanced melanoma. Here, apart from the molecular background of c-KIT and its cellular functions, we will review the wide distribution of alterations in KIT with a catalogue of more than 40 mutations reported in various articles and case studies. Additionally, we will summarize the association of KIT mutations with clinicopathologic features (age, sex, melanoma subtypes, anatomic location, etc.), and the differences of mutation rate among subgroups. Finally, several therapeutic trials of c-KIT inhibitors, including imatinib, dasatinib, nilotinib, and sunitinib, will be analyzed for their success rates and limitations in advanced melanoma treatment. These not only emphasize c-KIT as an attractive target for personalized melanoma therapy but also propose the requirement for additional investigational studies to develop novel therapeutic trials co-targeting c-KIT and other cytokines such as members of signaling pathways and immune systems.
Collapse
Affiliation(s)
- Duc Daniel M Pham
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, Korea
| | | | - Hensin Tsao
- Harvard Medical School, Boston, MA, USA
- Department of Dermatology, Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
24
|
Chen YC, Liao JW, Chang SC, Hsu WL. Expression frequency of c-kit isoforms and its prognostic potential in canine mammary tumours. Vet Comp Oncol 2019; 18:303-314. [PMID: 31652393 DOI: 10.1111/vco.12549] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Revised: 10/05/2019] [Accepted: 10/21/2019] [Indexed: 12/28/2022]
Abstract
KIT is a tyrosine kinase receptor involved in carcinogenesis. Two alternatively spliced transcripts, differed from presence of four amino acids (GNSK) at exon 9 of c-kit, were identified in various human tumours and canine hemangiosarcoma (HSA). The biological function and clinical implications of these isoforms have not yet been elucidated in canine tumours. The current study aimed to validate the expression profile and ultimately to evaluate the correlation of c-kit isoform levels with clinicopathological factors of canine mammary tumours (CMTs). In total, the expression profiles of c-kit isoforms in 196 samples obtained from normal mammary glands (NMGs) of healthy controls and dogs with CMTs, benign and malignant CMTs, and HSAs were determined by polymerase chain reaction (PCR) and quantified via real-time PCR. Overall, the expression levels of the two isoforms were equivalent in NMGs, whereas the GNSK- /GNSK+ ratio sharply increased to 7.44- and 8.33-fold, indicating abundant GNSK- isoforms in benign and malignant CMTs, respectively. However, a significant decrease in GNSK- expression was detected in dogs with high-grade malignant CMTs (mCMTs) and with metastatic CMTs compared with expression in those with a lower grade and non-metastatic CMTs. In addition, the median survival time was shorter in mCMT canines with a lower GNSK- /GNSK+ ratio than that in mCMT canines with a higher ratio (899 days vs 1534 days). In conclusion, two c-kit isoforms are ubiquitously expressed with great variability in HSAs and CMTs with both benign and malignant status. The GNSK- /GNSK+ ratio could serve as a prognostic indicator for dogs with mCMTs.
Collapse
Affiliation(s)
- Yi-Chen Chen
- Graduate Institute of Veterinary Pathobiology, College of Veterinary Medicine, National Chung Hsing University, Taichung, Taiwan
| | - Jiunn-Wang Liao
- Graduate Institute of Veterinary Pathobiology, College of Veterinary Medicine, National Chung Hsing University, Taichung, Taiwan
| | - Shih-Chieh Chang
- Department of Veterinary Medicine, College of Veterinary Medicine, National Chung Hsing University, Taichung, Taiwan.,Veterinary Medical Teaching Hospital, College of Veterinary Medicine, National Chung Hsing University, Taichung, Taiwan
| | - Wei-Li Hsu
- Graduate Institute of Microbiology and Public Health, College of Veterinary Medicine, National Chung Hsing University, Taichung, Taiwan
| |
Collapse
|
25
|
Vozdova M, Kubickova S, Fictum P, Cernohorska H, Fröhlich J, Rubes J. Mutation and methylation status of KIT and TP 53 in canine cutaneous and subcutaneous mast cell tumours. Vet Comp Oncol 2019; 18:438-444. [PMID: 31574575 DOI: 10.1111/vco.12543] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 09/10/2019] [Accepted: 09/16/2019] [Indexed: 01/08/2023]
Abstract
Cutaneous and subcutaneous mast cell tumours (MCTs) are counted among the most frequent cancers in dogs. However, the genetic aetiology of their development is still mostly unknown, with the exception of KIT and tumor protein p53 (TP53 ) mutations reported in less than a half of cutaneous MCTs. In subcutaneous MCTs, no gene alterations were previously detected. We analysed KIT and TP53 mutations in cutaneous and subcutaneous MCTs, and identified methylated CpG sites in KIT and TP53 promoters and adjacent exon 1 regions. The mutation analysis focused on KIT exons 8, 9 and 11, and TP53 exons 5-8, and revealed mutations in 26% and 7% cutaneous MCT cases, respectively. Moreover, we report a first case of KIT mutation ever detected in subcutaneous MCTs. KIT exon 11 mutations and high Kiupel and Patnaik grades were associated with reduced survival in this study. Both KIT and TP53 gene were generally unmethylated in canine cutaneous MCTs. A sporadic methylation of the CpG positions in KIT promoter and adjacent exon 1 was detected in 70.4% of cutaneous and 82% of subcutaneous MCTs. A sporadic methylation of the CpG positions in the TP53 promoter and exon 1 was observed in 36.8% of the analysed cutaneous MCT samples. Only in two subcutaneous MCTs, we observed more than 30% of clones showing KIT methylation at the CpG positions 13 or 14. The CpG position 14 is involved in a predicted binding site for Sp1 transcription factor. However, the significance of KIT promoter methylation at this specific position needs further evaluation.
Collapse
Affiliation(s)
- Miluse Vozdova
- Department of Genetics and Reproduction, Central European Institute of Technology-Veterinary Research Institute, Brno, Czech Republic
| | - Svatava Kubickova
- Department of Genetics and Reproduction, Central European Institute of Technology-Veterinary Research Institute, Brno, Czech Republic
| | - Petr Fictum
- Department of Pathological Morphology and Parasitology, Faculty of Veterinary Medicine, University of Veterinary and Pharmaceutical Sciences Brno, Brno, Czech Republic
| | - Halina Cernohorska
- Department of Genetics and Reproduction, Central European Institute of Technology-Veterinary Research Institute, Brno, Czech Republic
| | - Jan Fröhlich
- Department of Genetics and Reproduction, Central European Institute of Technology-Veterinary Research Institute, Brno, Czech Republic
| | - Jiri Rubes
- Department of Genetics and Reproduction, Central European Institute of Technology-Veterinary Research Institute, Brno, Czech Republic
| |
Collapse
|