1
|
Zhang S, Wang Y, Xiong X, Xing J, Jing K. Mechanistic insights into Hippo-YAP pathway activation for enhanced DFU healing. Am J Physiol Cell Physiol 2025; 328:C1921-C1940. [PMID: 40261295 DOI: 10.1152/ajpcell.01067.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Revised: 01/16/2025] [Accepted: 04/10/2025] [Indexed: 04/24/2025]
Abstract
With the increasing prevalence of diabetes, diabetic foot ulcers (DFUs) have become a global health challenge, significantly impacting patients' quality of life and placing a substantial burden on healthcare systems. Among various immune cell subsets, M2-polarized macrophages play a pivotal role in tissue repair and inflammation resolution. This study uses single-cell RNA sequencing (scRNA-seq) and bulk RNA sequencing to comprehensively investigate the role of the TFAP2A-LIFR-Hippo-YAP signaling axis in regulating macrophage M2 polarization and its critical function in DFU wound healing. Through scRNA-seq analysis, we identified nine major immune cell subsets in DFU samples, with macrophages emerging as key regulatory cells. In vitro experiments further confirmed that TFAP2A promotes macrophage M2 polarization (evidenced by increased expression of the M2 marker ARG1) and ameliorates endothelial dysfunction by enhancing tube formation, improving migration capacity, and upregulating relevant proteins such as VCAM-1. Moreover, TFAP2A serves as a central regulatory gene for macrophage function in DFU by upregulating LIFR expression and activating the Hippo-YAP signaling pathway, thereby inducing M2 polarization and mitigating endothelial dysfunction. Mouse model experiments further demonstrated that the TFAP2A-LIFR-Hippo-YAP signaling axis accelerates DFU wound healing through the induction of macrophage M2 polarization. This study unveils a novel immunoregulatory role of TFAP2A in DFU and provides a promising therapeutic target for the treatment of chronic diabetic wounds.NEW & NOTEWORTHY This study provides unprecedented insights into diabetic foot ulcer healing by demonstrating the novel immunoregulatory role of the TFAP2A-LIFR-Hippo-YAP signaling axis. Leveraging single-cell and bulk transcriptomics, we identify TFAP2A as a crucial regulator of macrophage M2 polarization, essential for wound healing and angiogenesis. These findings offer valuable mechanistic understanding and present TFAP2A as a promising therapeutic target for improving outcomes in chronic diabetic wounds.
Collapse
Affiliation(s)
- Shaochun Zhang
- Department of Orthopedics, The Central Hospital of Ezhou, Ezhou, People's Republic of China
| | - Ye Wang
- Department of Orthopedics, The Central Hospital of Ezhou, Ezhou, People's Republic of China
| | - Xuesong Xiong
- Department of Endocrinology, The Central Hospital of Ezhou, Ezhou, People's Republic of China
| | - Jili Xing
- Department of Gastroenterology, The Central Hospital of Ezhou, Ezhou, People's Republic of China
| | - Ke Jing
- Department of Endocrinology, The Central Hospital of Ezhou, Ezhou, People's Republic of China
| |
Collapse
|
2
|
Cui X, Yin Q, Gao Z, Li Z, Chen X, Lv H, Chen S, Liu Q, Zeng W, Jiang R. CREATE: cell-type-specific cis-regulatory element identification via discrete embedding. Nat Commun 2025; 16:4607. [PMID: 40382355 PMCID: PMC12085597 DOI: 10.1038/s41467-025-59780-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Accepted: 05/02/2025] [Indexed: 05/20/2025] Open
Abstract
Cis-regulatory elements (CREs), including enhancers, silencers, promoters and insulators, play pivotal roles in orchestrating gene regulatory mechanisms that drive complex biological traits. However, current approaches for CRE identification are predominantly sequence-based and typically focus on individual CRE types, limiting insights into their cell-type-specific functions and regulatory dynamics. Here, we present CREATE, a multimodal deep learning framework based on Vector Quantized Variational AutoEncoder, tailored for comprehensive CRE identification and characterization. CREATE integrates genomic sequences, chromatin accessibility, and chromatin interaction data to generate discrete CRE embeddings, enabling accurate multi-class classification and robust characterization of CREs. CREATE excels in identifying cell-type-specific CREs, and provides quantitative and interpretable insights into CRE-specific features, uncovering the underlying regulatory codes. By facilitating large-scale prediction of CREs in specific cell types, CREATE enhances the recognition of disease- or phenotype-associated biological variabilities of CREs, thus advancing our understanding of gene regulatory landscapes and their roles in health and disease.
Collapse
Affiliation(s)
- Xuejian Cui
- Ministry of Education Key Laboratory of Bioinformatics, Bioinformatics Division at the Beijing National Research Center for Information Science and Technology, Center for Synthetic and Systems Biology, Department of Automation, Tsinghua University, Beijing, China
| | - Qijin Yin
- Ministry of Education Key Laboratory of Bioinformatics, Bioinformatics Division at the Beijing National Research Center for Information Science and Technology, Center for Synthetic and Systems Biology, Department of Automation, Tsinghua University, Beijing, China
| | - Zijing Gao
- Ministry of Education Key Laboratory of Bioinformatics, Bioinformatics Division at the Beijing National Research Center for Information Science and Technology, Center for Synthetic and Systems Biology, Department of Automation, Tsinghua University, Beijing, China
| | - Zhen Li
- Ministry of Education Key Laboratory of Bioinformatics, Bioinformatics Division at the Beijing National Research Center for Information Science and Technology, Center for Synthetic and Systems Biology, Department of Automation, Tsinghua University, Beijing, China
| | - Xiaoyang Chen
- Ministry of Education Key Laboratory of Bioinformatics, Bioinformatics Division at the Beijing National Research Center for Information Science and Technology, Center for Synthetic and Systems Biology, Department of Automation, Tsinghua University, Beijing, China
| | - Hairong Lv
- Ministry of Education Key Laboratory of Bioinformatics, Bioinformatics Division at the Beijing National Research Center for Information Science and Technology, Center for Synthetic and Systems Biology, Department of Automation, Tsinghua University, Beijing, China
| | - Shengquan Chen
- School of Mathematical Sciences and LPMC, Nankai University, Tianjin, China
| | - Qiao Liu
- Department of Statistics, Stanford University, Stanford, CA, USA
| | - Wanwen Zeng
- Department of Statistics, Stanford University, Stanford, CA, USA.
| | - Rui Jiang
- Ministry of Education Key Laboratory of Bioinformatics, Bioinformatics Division at the Beijing National Research Center for Information Science and Technology, Center for Synthetic and Systems Biology, Department of Automation, Tsinghua University, Beijing, China.
| |
Collapse
|
3
|
de Carvalho LM, Carvalho VMA, Camargo AP, Papes F. Gene network analysis identifies dysregulated pathways in an autism spectrum disorder caused by mutations in Transcription Factor 4. Sci Rep 2025; 15:4993. [PMID: 39929970 PMCID: PMC11811132 DOI: 10.1038/s41598-025-89334-0] [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: 10/15/2024] [Accepted: 02/04/2025] [Indexed: 02/13/2025] Open
Abstract
Pitt-Hopkins syndrome (PTHS) is a rare neurodevelopmental monogenic disorder in the autistic spectrum caused by mutations in the Transcription Factor 4 gene. Even though the genetic etiology is known, the molecular mechanisms underlying PTHS remain poorly understood. To gain insight into the disease's pathophysiology, we set out to identify genes and pathways putatively involved in the pathology through co-expression and gene hub analyses using transcriptomic data from neural progenitor cells, neurons, and brain organoids derived from PTHS patients. Our results revealed several groups of co-expressed genes that are differentially regulated in PTHS neural cells compared to controls. These groups were enriched for genes involved in neural development and function, including synaptic transmission, membrane excitability, and cell adhesion. We identified several hub genes (highly connected nodes within gene networks that are central in these modules), including some that encode proteins involved in histone modification, synaptic vesicle trafficking, and cell signaling. Furthermore, we found that the differential expression of hub genes in PTHS neural cells was associated with altered cellular processes linked to neurodevelopment, such as cell-cell communication and irregular synaptic networks. Notably, we identified a set of hub genes related to the histone gene family, which is associated with neuronal differentiation and may contribute to PTHS pathogenesis and potentially serve as a biomarker for disease prognosis. Our results support the notion that PTHS involves alterations in neural development and function, particularly in excitatory neurons. The groups of co-expressed genes and hub genes we identified provide new insights into the molecular mechanisms underlying PTHS pathogenesis and could potentially be targeted for therapeutic intervention.
Collapse
Affiliation(s)
- Lucas M de Carvalho
- Department of Genetics, Evolution, Microbiology, and Immunology, Institute of Biology, University of Campinas, Campinas, Sao Paulo, 13083-862, Brazil.
- Sao Francisco University, Bragança Paulista, Sao Paulo, 12916-900, Brazil.
| | - Vinicius M A Carvalho
- Department of Genetics, Evolution, Microbiology, and Immunology, Institute of Biology, University of Campinas, Campinas, Sao Paulo, 13083-862, Brazil
- Department of Psychiatry and Behavioral Sciences, School of Medicine, Stanford University, Stanford, CA, 94305, USA
| | - Antonio P Camargo
- Department of Genetics, Evolution, Microbiology, and Immunology, Institute of Biology, University of Campinas, Campinas, Sao Paulo, 13083-862, Brazil.
- Lawrence Berkeley National Laboratory, Berkeley, CA, 94720, USA.
| | - Fabio Papes
- Department of Genetics, Evolution, Microbiology, and Immunology, Institute of Biology, University of Campinas, Campinas, Sao Paulo, 13083-862, Brazil.
- Department of Pediatrics, School of Medicine, University of California San Diego, La Jolla, CA, 92037, USA.
- Center for Medicinal Chemistry, University of Campinas, Campinas, Sao Paulo, 13083-886, Brazil.
| |
Collapse
|
4
|
Beck AC, Cho E, White JR, Paemka L, Li T, Gu VW, Thompson DT, Koch KE, Franke C, Gosse M, Wu VT, Landers SR, Pamatmat AJ, Kulak MV, Weigel RJ. AP-2α Regulates S-Phase and Is a Marker for Sensitivity to PI3K Inhibitor Buparlisib in Colon Cancer. Mol Cancer Res 2021; 19:1156-1167. [PMID: 33753551 DOI: 10.1158/1541-7786.mcr-20-0867] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 01/21/2021] [Accepted: 03/16/2021] [Indexed: 01/22/2023]
Abstract
Activating protein 2 alpha (AP-2α; encoded by TFAP2A) functions as a tumor suppressor and influences response to therapy in several cancer types. We aimed to characterize regulation of the transcriptome by AP-2α in colon cancer. CRISPR-Cas9 and short hairpin RNA were used to eliminate TFAP2A expression in HCT116 and a panel of colon cancer cell lines. AP-2α target genes were identified with RNA sequencing and chromatin immunoprecipitation sequencing. Effects on cell cycle were characterized in cells synchronized with aphidicolin and analyzed by FACS and Premo FUCCI. Effects on invasion and tumorigenesis were determined by invasion assay, growth of xenografts, and phosphorylated histone H3 (PHH3). Knockout of TFAP2A induced significant alterations in the transcriptome including repression of TGM2, identified as a primary gene target of AP-2α. Loss of AP-2α delayed progression through S-phase into G2-M and decreased phosphorylation of AKT, effects that were mediated through regulation of TGM2. Buparlisib (BKM120) repressed in vitro invasiveness of HCT116 and a panel of colon cancer cell lines; however, loss of AP-2α induced resistance to buparlisib. Similarly, buparlisib repressed PHH3 and growth of tumor xenografts and increased overall survival of tumor-bearing mice, whereas, loss of AP-2α induced resistance to the effect of PI3K inhibition. Loss of AP-2α in colon cancer leads to prolonged S-phase through altered activation of AKT leading to resistance to the PI3K inhibitor, Buparlisib. The findings demonstrate an important role for AP-2α in regulating progression through the cell cycle and indicates that AP-2α is a marker for response to PI3K inhibitors. IMPLICATIONS: AP-2α regulated cell cycle through the PI3K cascade and activation of AKT mediated through TGM2. AP-2α induced sensitivity to Buparlisib/BKM120, indicating that AP-2α is a biomarker predictive of response to PI3K inhibitors.
Collapse
Affiliation(s)
- Anna C Beck
- Department of Surgery, University of Iowa, Iowa City, Iowa
| | - Edward Cho
- Department of Surgery, University of Iowa, Iowa City, Iowa
| | | | - Lily Paemka
- Department of Surgery, University of Iowa, Iowa City, Iowa.,Department of Biochemistry, Cell and Molecular Biology, West African Center for Cell Biology of Infectious Pathogens, School of Biological Sciences, College of Basic and Applied Science University of Ghana, Accra, Ghana
| | - Tiandao Li
- Department of Surgery, University of Iowa, Iowa City, Iowa
| | - Vivian W Gu
- Department of Surgery, University of Iowa, Iowa City, Iowa
| | | | - Kelsey E Koch
- Department of Surgery, University of Iowa, Iowa City, Iowa
| | | | - Matthew Gosse
- Department of Pathology, University of Iowa, Iowa City, Iowa
| | - Vincent T Wu
- Department of Surgery, University of Iowa, Iowa City, Iowa
| | | | | | | | | |
Collapse
|
5
|
Shi S, Huang X, Ma X, Zhu X, Zhang Q. Research of the mechanism on miRNA193 in exosomes promotes cisplatin resistance in esophageal cancer cells. PLoS One 2020; 15:e0225290. [PMID: 32369495 PMCID: PMC7199973 DOI: 10.1371/journal.pone.0225290] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Accepted: 01/30/2020] [Indexed: 02/06/2023] Open
Abstract
Purpose Chemotherapy resistance of esophageal cancer is a key factor affecting the postoperative treatment of esophageal cancer. Among the media that transmit signals between cells, the exosomes secreted by tumor cells mediate information transmission between tumor cells, which can make sensitive cells obtain resistance. Although some cellular exosomes play an important role in tumor’s acquired drug resistance, the related action mechanism is still not explored specifically. Methods To elucidate this process, we constructed a cisplatin-resistant esophageal cancer cell line, and proved that exosomes conferring cellular resistance in esophageal cancer can promote cisplatin resistance in sensitive cells. Through high-throughput sequencing analysis of the exosome and of cells after stimulation by exosomes, we determined that the miRNA193 in exosomes conferring cellular resistance played a key role in sensitive cells acquiring resistance to cisplatin. In vitro experiments showed that miRNA193 can regulate the cell cycle of esophageal cancer cells and inhibit apoptosis, so that sensitive cells can acquire resistance to cisplatin. An in vivo experiment proved that miRNA193 can promote tumor proliferation through the exosomes, and provide sensitive cells with slight resistance to cisplatin. Results Small RNA sequencing of exosomes showed that exosomes in drug-resistant cells have 189 up-regulated and 304 down-regulated miRNAs; transcriptome results showed that drug-sensitive cells treated with drug-resistant cellular exosomes have 3446 high-expression and 1709 low-expression genes; correlation analysis showed that drug-resistant cellular exosomes mainly affect the drug resistance of sensitive cells through paths such as cytokine–cytokine receptor interaction, and the VEGF and Jak-STAT signaling pathways; miRNA193, one of the high-expression miRNAs in drug-resistant cellular exosomes, can promote drug resistance by removing cisplatin’s inhibition of the cell cycle of sensitive cells. Conclusion Sensitive cells can become resistant to cisplatin through acquired drug-resistant cellular exosomes, and miRNA193 can make tumor cells acquire cisplatin resistance by regulating the cell cycle.
Collapse
Affiliation(s)
- Shifeng Shi
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
- SanQuan Medical College, Xinxiang Medical University, Xinxiang, China
| | - Xin Huang
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Xiao Ma
- The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Xiaoyan Zhu
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
- * E-mail: (XZ); (QZ)
| | - Qinxian Zhang
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
- * E-mail: (XZ); (QZ)
| |
Collapse
|
6
|
Ross GR, Edwards S, Warner C, Homar P, Downey FX, Emelyanova L, Rizvi F, Jahangir A. Deletion of transcription factor AP-2α gene attenuates fibroblast differentiation into myofibroblast. J Cell Mol Med 2019; 23:6494-6498. [PMID: 31339227 PMCID: PMC6714505 DOI: 10.1111/jcmm.14421] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Revised: 03/26/2019] [Accepted: 04/18/2019] [Indexed: 12/18/2022] Open
Affiliation(s)
- Gracious R Ross
- Center for Integrative Research on Cardiovascular Aging, Aurora Research Institute, Aurora Health Care, Milwaukee, Wisconsin
| | - Stacie Edwards
- Center for Integrative Research on Cardiovascular Aging, Aurora Research Institute, Aurora Health Care, Milwaukee, Wisconsin
| | - Catherine Warner
- Center for Integrative Research on Cardiovascular Aging, Aurora Research Institute, Aurora Health Care, Milwaukee, Wisconsin
| | - Peter Homar
- Center for Integrative Research on Cardiovascular Aging, Aurora Research Institute, Aurora Health Care, Milwaukee, Wisconsin
| | - Francis X Downey
- Aurora Cardiovascular Services, Aurora Sinai/Aurora St. Luke's Medical Centers, Milwaukee, Wisconsin
| | - Larisa Emelyanova
- Center for Integrative Research on Cardiovascular Aging, Aurora Research Institute, Aurora Health Care, Milwaukee, Wisconsin
| | - Farhan Rizvi
- Center for Integrative Research on Cardiovascular Aging, Aurora Research Institute, Aurora Health Care, Milwaukee, Wisconsin
| | - Arshad Jahangir
- Aurora Cardiovascular Services, Aurora Sinai/Aurora St. Luke's Medical Centers, Milwaukee, Wisconsin
| |
Collapse
|
7
|
Kołat D, Kałuzińska Ż, Bednarek AK, Płuciennik E. The biological characteristics of transcription factors AP-2α and AP-2γ and their importance in various types of cancers. Biosci Rep 2019; 39:BSR20181928. [PMID: 30824562 PMCID: PMC6418405 DOI: 10.1042/bsr20181928] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Revised: 02/11/2019] [Accepted: 02/27/2019] [Indexed: 02/07/2023] Open
Abstract
The Activator Protein 2 (AP-2) transcription factor (TF) family is vital for the regulation of gene expression during early development as well as carcinogenesis process. The review focusses on the AP-2α and AP-2γ proteins and their dualistic regulation of gene expression in the process of carcinogenesis. Both AP-2α and AP-2γ influence a wide range of physiological or pathological processes by regulating different pathways and interacting with diverse molecules, i.e. other proteins, long non-coding RNAs (lncRNA) or miRNAs. This review summarizes the newest information about the biology of two, AP-2α and AP-2γ, TFs in the carcinogenesis process. We emphasize that these two proteins could have either oncogenic or suppressive characteristics depending on the type of cancer tissue or their interaction with specific molecules. They have also been found to contribute to resistance and sensitivity to chemotherapy in oncological patients. A better understanding of molecular network of AP-2 factors and other molecules may clarify the atypical molecular mechanisms occurring during carcinogenesis, and may assist in the recognition of new diagnostic biomarkers.
Collapse
Affiliation(s)
- Damian Kołat
- Faculty of Biomedical Sciences and Postgraduate Education, Medical University of Lodz, Lodz, Poland
| | - Żaneta Kałuzińska
- Faculty of Biomedical Sciences and Postgraduate Education, Medical University of Lodz, Lodz, Poland
| | - Andrzej K Bednarek
- Department of Molecular Carcinogenesis, Medical University of Lodz, Lodz, Poland
| | - Elżbieta Płuciennik
- Department of Molecular Carcinogenesis, Medical University of Lodz, Lodz, Poland
| |
Collapse
|
8
|
Huang D, Petrykowska HM, Miller BF, Elnitski L, Ovcharenko I. Identification of human silencers by correlating cross-tissue epigenetic profiles and gene expression. Genome Res 2019; 29:657-667. [PMID: 30886051 PMCID: PMC6442386 DOI: 10.1101/gr.247007.118] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Accepted: 02/14/2019] [Indexed: 12/22/2022]
Abstract
Compared to enhancers, silencers are notably difficult to identify and validate experimentally. In search for human silencers, we utilized H3K27me3-DNase I hypersensitive site (DHS) peaks with tissue specificity negatively correlated with the expression of nearby genes across 25 diverse cell lines. These regions are predicted to be silencers since they are physically linked, using Hi-C loops, or associated, using expression quantitative trait loci (eQTL) results, with a decrease in gene expression much more frequently than general H3K27me3-DHSs. Also, these regions are enriched for the binding sites of transcriptional repressors (such as CTCF, MECOM, SMAD4, and SNAI3) and depleted of the binding sites of transcriptional activators. Using sequence signatures of these regions, we constructed a computational model and predicted approximately 10,000 additional silencers per cell line and demonstrated that the majority of genes linked to these silencers are expressed at a decreased level. Furthermore, single nucleotide polymorphisms (SNPs) in predicted silencers are significantly associated with disease phenotypes. Finally, our results show that silencers commonly interact with enhancers to affect the transcriptional dynamics of tissue-specific genes and to facilitate fine-tuning of transcription in the human genome.
Collapse
Affiliation(s)
- Di Huang
- Computational Biology Branch, National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Hanna M Petrykowska
- Translational and Functional Genomics Branch, National Human Genome Research, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Brendan F Miller
- Translational and Functional Genomics Branch, National Human Genome Research, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Laura Elnitski
- Translational and Functional Genomics Branch, National Human Genome Research, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Ivan Ovcharenko
- Computational Biology Branch, National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, Maryland 20892, USA
| |
Collapse
|
9
|
Feng X, Han H, Zou D, Zhou J, Zhou W. Suberoylanilide hydroxamic acid-induced specific epigenetic regulation controls Leptin-induced proliferation of breast cancer cell lines. Oncotarget 2018; 8:3364-3379. [PMID: 27926517 PMCID: PMC5356888 DOI: 10.18632/oncotarget.13764] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Accepted: 11/22/2016] [Indexed: 11/25/2022] Open
Abstract
Breast cancer is one of the most common malignancies among women in the world, investigating the characteristics and special transduction pathways is important for better understanding breast development and tumorigenesis. Leptin, a peptide hormone secreted from white adipocytes, may be an independent risk factor for breast cancer. Here, we treated suberoylanilide hydroxamic acid (SAHA) on Leptin-induced cell proliferation and invasion in the estrogen-receptor-positive breast cancer cell line MCF-7 and triple-negative breast cancer cell line MDA-MB-231. Low concentrations of Leptin (0.625 nM) significantly stimulated breast cancer cell growth, enhanced cell viability, minimized apoptosis, and increased cell cycle transition. In contrast, SAHA (5 μM) treatment had reverse effects. Wound healing assay showed that, in MCF-7 and MDA-MB-231 cell line, cell migrating stimulated by Leptin was significantly repressed with SAHA treatment. Moreover, cell cycle real-time PCR array and proteome profiler antibody array confirmed that Leptin and SAHA treatment significantly changed the expressions of factors associated with cell cycle regulation and apoptosis including p53 and p21WAF1/CIP1. In DNA-ChIP analysis, we found that acetylation levels binding with p21WAF1/CIP1 promoters are regulated in a manner specific to histone type, lysine residue and selective promoter regions. SAHA significantly up-regulated the acetylation levels of AcH3-k14 and AcH3-k27 in MCF-7 cells, whereas Leptin repressed the modification. In addition, SAHA or Leptin had no significant effects on the AcH4 acetylation binding with any regions of p21WAF1/CIP1 promoter. In MDA-MB-231 cells, SAHA alone or in combination with Leptin significantly increased acetylation levels of Ach3-k27, Ach3-k18 and Ach4-k5 residues. However, no clear change was found with Leptin alone at all. Overall, our data will inform future studies to elucidate the mechanisms of p21WAF1/CIP1 transcriptional regulation, and the functional roles of p21WAF1/CIP1 in breast cancer tumorigenesis.
Collapse
Affiliation(s)
- Xiuyan Feng
- The Second Affiliated Hospital of Shenyang Medical College, Heping District, Shenyang City, Liaoning Province 110002, P. R. China.,Key Laboratory of Environmental Pollution and Microecology of Liaoning Province, Shenyang Medical College, Huanggu District, Shenyang City, Liaoning Province 110034, P. R. China
| | - Han Han
- Key Laboratory of Environmental Pollution and Microecology of Liaoning Province, Shenyang Medical College, Huanggu District, Shenyang City, Liaoning Province 110034, P. R. China
| | - Dan Zou
- Key Laboratory of Environmental Pollution and Microecology of Liaoning Province, Shenyang Medical College, Huanggu District, Shenyang City, Liaoning Province 110034, P. R. China
| | - Jiaming Zhou
- Northeast Yucai Foreign Language School, Hunnan New District, Shenyang City, Liaoning Province 110179, P. R. China
| | - Weiqiang Zhou
- Key Laboratory of Environmental Pollution and Microecology of Liaoning Province, Shenyang Medical College, Huanggu District, Shenyang City, Liaoning Province 110034, P. R. China
| |
Collapse
|
10
|
Kousa YA, Mansour TA, Seada H, Matoo S, Schutte BC. Shared molecular networks in orofacial and neural tube development. Birth Defects Res 2017; 109:169-179. [DOI: 10.1002/bdra.23598] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2016] [Revised: 10/17/2016] [Accepted: 10/21/2016] [Indexed: 11/06/2022]
Affiliation(s)
- Youssef A. Kousa
- Pediatric Residency Program; Children's National Health System; Washington DC
| | - Tamer A. Mansour
- Department of Population Health and Reproduction; University of California; Davis California
- Department of Clinical Pathology, College of Medicine; Mansoura University; Egypt
| | - Haitham Seada
- Department of Computer Science and Engineering, Computational Optimization and Innovation Laboratory; Michigan State University; East Lansing Michigan
| | - Samaneh Matoo
- Department of Modern Science; Islamic Azad University-Tehran Medical Branch; Tehran Iran
| | - Brian C. Schutte
- Department of Microbiology and Molecular Genetics and the Department of Pediatrics and Human Development; Michigan State University; East Lansing Michigan
| |
Collapse
|
11
|
Zhou J, Duan H, Xie Y, Ning Y, Zhang X, Hui N, Wang C, Zhang J, Zhou J. MiR-193a-5p Targets the Coding Region of AP-2α mRNA and Induces Cisplatin Resistance in Bladder Cancers. J Cancer 2016; 7:1740-1746. [PMID: 27698912 PMCID: PMC5039396 DOI: 10.7150/jca.15620] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Accepted: 06/29/2016] [Indexed: 12/21/2022] Open
Abstract
Transcription factor AP-2 alpha (AP-2α or TFAP2A) is a newly identified prognostic marker of chemotherapy; its expression is positively correlated with chemosensitivity and survival of cancer patients. Using computational programs, we predicted that the coding region of AP-2α gene contains a potential miRNA response element (MRE) of miR-193a-5p, and the single nucleotide polymorphism (SNP) site (c.497A>G, rs111681798) resides within the predicted MRE. The results of luciferase assays and Western blot analysis demonstrated that miR-193a-5p negatively regulated the expression of AP-2α proteins, but have no influence on the mutant AP-2α (c.497A>G). Infection with lentiviral AP-2α gene or miR-193a-5p inhibitor in the bladder cancer cells decreased migration and cisplatin resistance, while knockdown of AP-2α gene or overexpression of miR-193a-5p in the urothelial cell line SV-HUC-1 increased migration and cisplatin resistances. We concluded that miR-193a-5p induced cisplatin resistance by repressing AP-2α expression in bladder cancer cells.
Collapse
Affiliation(s)
- Ji Zhou
- College of Life Science, Hunan Normal University, Changsha 410081, Hunan, China
| | - Huaxin Duan
- Hunan Provincial People's Hospital, Changsha 410005, Hunan, China
| | - Yu Xie
- Hunan Cancer Hospital, Changsha 410013, Hunan, China
| | - Yichong Ning
- College of Life Science, Hunan Normal University, Changsha 410081, Hunan, China
| | - Xing Zhang
- College of Life Science, Hunan Normal University, Changsha 410081, Hunan, China
| | - Na Hui
- College of Life Science, Hunan Normal University, Changsha 410081, Hunan, China
| | - Chunqing Wang
- College of Life Science, Hunan Normal University, Changsha 410081, Hunan, China
| | - Jian Zhang
- College of Life Science, Hunan Normal University, Changsha 410081, Hunan, China
| | - Jianlin Zhou
- College of Life Science, Hunan Normal University, Changsha 410081, Hunan, China
| |
Collapse
|
12
|
Rondón-Lagos M, Rangel N, Di Cantogno LV, Annaratone L, Castellano I, Russo R, Manetta T, Marchiò C, Sapino A. Effect of low doses of estradiol and tamoxifen on breast cancer cell karyotypes. Endocr Relat Cancer 2016; 23:635-50. [PMID: 27357940 PMCID: PMC5064758 DOI: 10.1530/erc-16-0078] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Accepted: 06/29/2016] [Indexed: 12/21/2022]
Abstract
Evidence supports a role of 17&-estradiol (E2) in carcinogenesis and the large majority of breast carcinomas are dependent on estrogen. The anti-estrogen tamoxifen (TAM) is widely used for both treatment and prevention of breast cancer; however, it is also carcinogenic in human uterus and rat liver, highlighting the profound complexity of its actions. The nature of E2- or TAM-induced chromosomal damage has been explored using relatively high concentrations of these agents, and only some numerical aberrations and chromosomal breaks have been analyzed. This study aimed to determine the effects of low doses of E2 and TAM (10(&8 )mol L(&1) and 10(&6 )mol L(&1) respectively) on karyotypes of MCF7, T47D, BT474, and SKBR3 breast cancer cells by comparing the results of conventional karyotyping and multi-FISH painting with cell proliferation. Estrogen receptor (ER)-positive (+) cells showed an increase in cell proliferation after E2 treatment (MCF7, T47D, and BT474) and a decrease after TAM treatment (MCF7 and T47D), whereas in ER& cells (SKBR3), no alterations in cell proliferation were observed, except for a small increase at 96 h. Karyotypes of both ER+ and ER& breast cancer cells increased in complexity after treatments with E2 and TAM leading to specific chromosomal abnormalities, some of which were consistent throughout the treatment duration. This genotoxic effect was higher in HER2+ cells. The ER&/HER2+ SKBR3 cells were found to be sensitive to TAM, exhibiting an increase in chromosomal aberrations. These in vitro results provide insights into the potential role of low doses of E2 and TAM in inducing chromosomal rearrangements in breast cancer cells.
Collapse
Affiliation(s)
| | - Nelson Rangel
- Department of Medical SciencesUniversity of Turin, Turin, Italy Natural and Mathematical Sciences FacultyUniversidad del Rosario, Bogotá, Colombia
| | | | | | | | - Rosalia Russo
- Department of Medical SciencesUniversity of Turin, Turin, Italy
| | - Tilde Manetta
- Department of Public Health and PediatricsUniversity of Turin, Turin, Italy
| | | | - Anna Sapino
- Department of Medical SciencesUniversity of Turin, Turin, Italy Candiolo Cancer InstituteFPO-IRCCS, Candiolo, Italy
| |
Collapse
|
13
|
Yoldi G, Pellegrini P, Trinidad EM, Cordero A, Gomez-Miragaya J, Serra-Musach J, Dougall WC, Muñoz P, Pujana MA, Planelles L, González-Suárez E. RANK Signaling Blockade Reduces Breast Cancer Recurrence by Inducing Tumor Cell Differentiation. Cancer Res 2016; 76:5857-5869. [PMID: 27480274 DOI: 10.1158/0008-5472.can-15-2745] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Accepted: 07/07/2016] [Indexed: 11/16/2022]
Abstract
RANK expression is associated with poor prognosis in breast cancer even though its therapeutic potential remains unknown. RANKL and its receptor RANK are downstream effectors of the progesterone signaling pathway. However, RANK expression is enriched in hormone receptor negative adenocarcinomas, suggesting additional roles for RANK signaling beyond its hormone-dependent function. Here, to explore the role of RANK signaling once tumors have developed, we use the mouse mammary tumor virus-Polyoma Middle T (MMTV-PyMT), which mimics RANK and RANKL expression patterns seen in human breast adenocarcinomas. Complementary genetic and pharmacologic approaches demonstrate that therapeutic inhibition of RANK signaling drastically reduces the cancer stem cell pool, decreases tumor and metastasis initiation, and enhances sensitivity to chemotherapy. Mechanistically, genome-wide expression analyses show that anti-RANKL therapy promotes lactogenic differentiation of tumor cells. Moreover, RANK signaling in tumor cells negatively regulates the expression of Ap2 transcription factors, and enhances the Wnt agonist Rspo1 and the Sca1-population, enriched in tumor-initiating cells. In addition, we found that expression of TFAP2B and the RANK inhibitor, OPG, in human breast cancer correlate and are associated with relapse-free tumors. These results support the use of RANKL inhibitors to reduce recurrence and metastasis in breast cancer patients based on its ability to induce tumor cell differentiation. Cancer Res; 76(19); 5857-69. ©2016 AACR.
Collapse
Affiliation(s)
- Guillermo Yoldi
- Cancer Epigenetics and Biology Program, Bellvitge Biomedical Research Institute, IDIBELL, Barcelona, Spain
| | - Pasquale Pellegrini
- Cancer Epigenetics and Biology Program, Bellvitge Biomedical Research Institute, IDIBELL, Barcelona, Spain
| | - Eva M Trinidad
- Cancer Epigenetics and Biology Program, Bellvitge Biomedical Research Institute, IDIBELL, Barcelona, Spain
| | - Alex Cordero
- Cancer Epigenetics and Biology Program, Bellvitge Biomedical Research Institute, IDIBELL, Barcelona, Spain
| | - Jorge Gomez-Miragaya
- Cancer Epigenetics and Biology Program, Bellvitge Biomedical Research Institute, IDIBELL, Barcelona, Spain
| | - Jordi Serra-Musach
- Program Against Cancer Therapeutic Resistance (ProCURE), Breast Cancer and Systems Biology Lab, Catalan Institute of Oncology, IDIBELL, Barcelona, Spain
| | | | - Purificación Muñoz
- Cancer Epigenetics and Biology Program, Bellvitge Biomedical Research Institute, IDIBELL, Barcelona, Spain
| | - Miguel-Angel Pujana
- Program Against Cancer Therapeutic Resistance (ProCURE), Breast Cancer and Systems Biology Lab, Catalan Institute of Oncology, IDIBELL, Barcelona, Spain
| | - Lourdes Planelles
- Centro Nacional de Biotecnología/CSIC, UAM Cantoblanco, Madrid, Spain
| | - Eva González-Suárez
- Cancer Epigenetics and Biology Program, Bellvitge Biomedical Research Institute, IDIBELL, Barcelona, Spain.
| |
Collapse
|
14
|
Marneros AG. Genetics of Aplasia Cutis Reveal Novel Regulators of Skin Morphogenesis. J Invest Dermatol 2015; 135:666-672. [DOI: 10.1038/jid.2014.413] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2014] [Revised: 09/01/2014] [Accepted: 09/04/2014] [Indexed: 11/09/2022]
|
15
|
Bogachek MV, Chen Y, Kulak MV, Woodfield GW, Cyr AR, Park JM, Spanheimer PM, Li Y, Li T, Weigel RJ. Sumoylation pathway is required to maintain the basal breast cancer subtype. Cancer Cell 2014; 25:748-61. [PMID: 24835590 PMCID: PMC4096794 DOI: 10.1016/j.ccr.2014.04.008] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2013] [Revised: 11/12/2013] [Accepted: 04/11/2014] [Indexed: 01/28/2023]
Abstract
The TFAP2C/AP-2γ transcription factor regulates luminal breast cancer genes, and loss of TFAP2C induces epithelial-mesenchymal transition. By contrast, the highly homologous family member, TFAP2A, lacks transcriptional activity at luminal gene promoters. A detailed structure-function analysis identified that sumoylation of TFAP2A blocks its ability to induce the expression of luminal genes. Disruption of the sumoylation pathway by knockdown of sumoylation enzymes, mutation of the SUMO-target lysine of TFAP2A, or treatment with sumoylation inhibitors induced a basal-to-luminal transition, which was dependent on TFAP2A. Sumoylation inhibitors cleared the CD44(+/hi)/CD24(-/low) cell population characterizing basal cancers and inhibited tumor outgrowth of basal cancer xenografts. These findings establish a critical role for sumoylation in regulating the transcriptional mechanisms that maintain the basal cancer phenotype.
Collapse
Affiliation(s)
- Maria V Bogachek
- Department of Surgery, University of Iowa, Iowa City, IA 52242, USA
| | - Yizhen Chen
- Department of Surgery, University of Iowa, Iowa City, IA 52242, USA
| | - Mikhail V Kulak
- Department of Surgery, University of Iowa, Iowa City, IA 52242, USA
| | | | - Anthony R Cyr
- Department of Surgery, University of Iowa, Iowa City, IA 52242, USA
| | - Jung M Park
- Department of Anatomy and Cell Biology, University of Iowa, Iowa City, IA 52242, USA
| | | | - Yingyue Li
- Department of Surgery, University of Iowa, Iowa City, IA 52242, USA
| | - Tiandao Li
- Department of Surgery, University of Iowa, Iowa City, IA 52242, USA; The Genome Institute, Washington University in St. Louis, St. Louis, MO 63108, USA
| | - Ronald J Weigel
- Department of Surgery, University of Iowa, Iowa City, IA 52242, USA; Department of Anatomy and Cell Biology, University of Iowa, Iowa City, IA 52242, USA; Department of Biochemistry, University of Iowa, Iowa City, IA 52242, USA.
| |
Collapse
|
16
|
Foertsch F, Teichmann N, Kob R, Hentschel J, Laubscher U, Melle C. S100A11 is involved in the regulation of the stability of cell cycle regulator p21(CIP1/WAF1) in human keratinocyte HaCaT cells. FEBS J 2013; 280:3840-53. [PMID: 23745637 DOI: 10.1111/febs.12378] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2013] [Revised: 06/03/2013] [Accepted: 06/04/2013] [Indexed: 01/13/2023]
Abstract
The cyclin-dependent kinase inhibitor p21(CIP1/WAF1) is a regulatory factor of the cell cycle. Its transcriptional activation and protein stability are tightly controlled by several distinct mechanisms. S100A11 is a member of the S100 family of Ca²⁺-binding proteins involved in several biological processes, including cell cycle progression and signal transduction. In the present study, we show that down-regulation of S100A11 results in the reduction of p21 protein in human HaCaT keratinocytes. It appears that a ubiquitin-independent proteasomal degradation process is involved in p21 degradation in S100A11 down-regulated cells. The application of a proteasome inhibitor stabilized p21 protein in these cells. Analysis of distinct signal transduction pathways revealed a disturbed phosphatidylinositol-3-kinase/Akt pathway after S100A11 knockdown. We determined that the glycogen synthase kinase-3, which is negatively regulated by phosphatidylinositol 3-kinase/Akt, was activated in cells possessing knocked-down S100A11 and appears to be involved in p21 protein destabilization. The application of a specific inhibitor of glycogen synthase kinase 3 resulted in an increase of the p21 protein level in S100A11 down-regulated HaCaT cells. Glycogen synthase kinase 3 is able to phosphorylate p21 at T57, which induces p21 proteasomal turnover. Mutation of the glycogen synthase kinase 3 site threonine 57 into alanine (T57A) stabilizes p21 in HaCaT cells lacking S100A11. Beside decreased p21 protein, down-regulation of S100A11 triggered the induction of apoptosis in HaCaT cells. These observations suggest that S100A11 is involved in the maintenance of p21 protein stability and appears to function as an inhibitor of apoptosis in human HaCaT keratinocyte cells. Thus, the data shed light on a novel pathway regulating p21 protein stability.
Collapse
|
17
|
Transcriptional regulation of the GPX1 gene by TFAP2C and aberrant CpG methylation in human breast cancer. Oncogene 2012; 32:4043-51. [PMID: 22964634 PMCID: PMC3522755 DOI: 10.1038/onc.2012.400] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2012] [Revised: 07/16/2012] [Accepted: 07/17/2012] [Indexed: 12/11/2022]
Abstract
The complexity of gene regulation has created obstacles to defining mechanisms that establish the patterns of gene expression characteristic of the different clinical phenotypes of breast cancer. TFAP2C is a transcription factor that has a critical role in the regulation of both estrogen receptor-alpha (ERα) and c-ErbB2/HER2 (Her2). Herein, we performed chromatin immunoprecipitation and direct sequencing (ChIP-seq) for TFAP2C in four breast cancer cell lines. Comparing the genomic binding sites for TFAP2C, we identified that glutathione peroxidase (GPX1) is regulated by TFAP2C through an AP-2 regulatory region in the promoter of the GPX1 gene. Knockdown of TFAP2C, but not the related factor TFAP2A, resulted in an abrogation of GPX1 expression. Selenium-dependent GPX activity correlated with endogenous GPX1 expression and overexpression of exogenous GPX1 induced GPX activity and significantly increased resistance to tert-butyl hydroperoxide. Methylation of the CpG island encompassing the AP-2 regulatory region was identified in cell lines where TFAP2C failed to bind the GPX1 promoter and GPX1 expression was unresponsive to TFAP2C. Furthermore, in cell lines where GPX1 promoter methylation was associated with gene silencing, treatment with 5'-aza-2-deoxycytidine (5'-aza-dC) (an inhibitor of DNA methylation) allowed TFAP2C to bind to the GPX1 promoter resulting in the activation of GPX1 RNA and protein expression. Methylation of the GPX1 promoter was identified in ∼20% of primary breast cancers and a highly significant correlation between the TFAP2C and GPX1 expression was confirmed when considering only those tumors with an unmethylated promoter, whereas the related factor, TFAP2A, failed to demonstrate a correlation. The results demonstrate that TFAP2C regulates the expression of GPX1, which influences the redox state and sensitivity to oxidative stress induced by peroxides. Given the established role of GPX1 in breast cancer, the results provide an important mechanism for TFAP2C to further influence oncogenesis and progression of breast carcinoma cells.
Collapse
|
18
|
Histone demethylase KDM5B collaborates with TFAP2C and Myc to repress the cell cycle inhibitor p21(cip) (CDKN1A). Mol Cell Biol 2012; 32:1633-44. [PMID: 22371483 DOI: 10.1128/mcb.06373-11] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The TFAP2C transcription factor has been shown to downregulate transcription of the universal cell cycle inhibitor p21(cip) (CDKN1A). In examining the mechanism of TFAP2C-mediated repression, we have identified a ternary complex at the proximal promoter containing TFAP2C, the oncoprotein Myc, and the trimethylated lysine 4 of histone H3 (H3K4me3) demethylase, KDM5B. We demonstrated that while TFAP2C and Myc can downregulate the CDKN1A promoter independently, KDM5B acts as a corepressor dependent on the other two proteins. All three factors collaborate for optimal CDKN1A repression, which requires the AP-2 binding site at -111/-103 and KDM5B demethylase activity. Silencing of TFAP2C-KDM5B-Myc led to increased H3K4me3 at the endogenous promoter and full induction of CDKN1A expression. Coimmunoprecipitation assays showed that TFAP2C and Myc associate with distinct domains of KDM5B and the TFAP2C C-terminal 270 amino acids (aa) are required for Myc and KDM5B interaction. Overexpression of all three proteins resulted in forced S-phase entry and attenuation of checkpoint activation, even in the presence of chemotherapy drugs. Since each protein has been linked to poor prognosis in breast cancer, our findings suggest that the TFAP2C-Myc-KDM5B complex promotes cell cycle progression via direct CDKN1A repression, thereby contributing to tumorigenesis and therapy failure.
Collapse
|
19
|
Rhodes JD, Lott MC, Russell SL, Moulton V, Sanderson J, Wormstone IM, Broadway DC. Activation of the innate immune response and interferon signalling in myotonic dystrophy type 1 and type 2 cataracts. Hum Mol Genet 2011; 21:852-62. [DOI: 10.1093/hmg/ddr515] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
|