1
|
Bounder G, Jouimyi MR, Essaidi I, Elyounsi I, Boura H, Michel V, Badre W, Touati E, Maachi F. Upstream stimulating factor 1 (USF1) -202 G/A polymorphism and serum levels of USF1 and USF2 are associated with gastric cancer risk: a case control study. J Cancer Res Clin Oncol 2025; 151:113. [PMID: 40102295 PMCID: PMC11919976 DOI: 10.1007/s00432-025-06158-1] [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: 01/07/2025] [Accepted: 03/04/2025] [Indexed: 03/20/2025]
Abstract
PURPOSE Gastric cancer is an inflammation-driven disease often associated with a bad prognosis. Upstream stimulatory factors USF1 and USF2 are pleiotropic transcription factors, with tumor suppressor function. Low expression of USF1 is associated with low survival in gastric cancer patients. USF1 genetic polymorphism -202G > A has been associated with cancer susceptibility. Our aim was to investigate USF1 gene polymorphism and serum level with the risk of gastric cancer. METHODS USF1-202 G/A polymorphism was analyzed by sanger sequencing, with the measure of USF1/USF2 serum levels by ELISA in H. pylori-positive patients with chronic gastritis, gastric precancerous lesions, gastric cancer and in healthy controls. RESULTS Our results show that the presence of the USF1-202 A allele increased the risk of gastric cancer compared to G (OR = 2; 95% CI 1.07-3.9; P = 0.02). Genotypically and under the dominant mutation model, the combined USF1- GA/AA -202 genotypes corresponded to higher risk of gastric cancer (OR = 3.5; 95% CI 1.4-8.2; p-value = 0.005) than the GG genotype. Moreover, the G/A transition at USF1-202 was associated with lower USF1 serum level, and mostly observed in gastric cancer patients where the average serological level of USF1 were 2.3 and twofold lower for the AA and GA genotypes, respectively, compared to GG. CONCLUSION USF1-202 G/A polymorphism constitutes a gastric cancer genetic risk factor. Together with USF1/USF2 serum level, they can be proposed as promising biomarkers for gastric cancer detection/prevention.
Collapse
Grants
- ACIP2015-10 INSTITUT PASTEUR Paris, as a Pasteur International Concerted action,
- ACIP2015-10 INSTITUT PASTEUR Paris, as a Pasteur International Concerted action,
- ACIP2015-10 INSTITUT PASTEUR Paris, as a Pasteur International Concerted action,
- ACIP2015-10 INSTITUT PASTEUR Paris, as a Pasteur International Concerted action,
- ACIP2015-10 INSTITUT PASTEUR Paris, as a Pasteur International Concerted action,
- ACIP2015-10 INSTITUT PASTEUR Paris, as a Pasteur International Concerted action,
- ACIP2015-10 INSTITUT PASTEUR Paris, as a Pasteur International Concerted action,
- Institut Pasteur du Maroc
Collapse
Affiliation(s)
- Ghizlane Bounder
- Helicobacter Pylori and Gastric Pathologies Laboratory, Institut Pasteur du Maroc, Casablanca, Morocco
| | - Mohamed Reda Jouimyi
- Helicobacter Pylori and Gastric Pathologies Laboratory, Institut Pasteur du Maroc, Casablanca, Morocco
| | - Imane Essaidi
- Helicobacter Pylori and Gastric Pathologies Laboratory, Institut Pasteur du Maroc, Casablanca, Morocco
| | | | - Hasna Boura
- Helicobacter Pylori and Gastric Pathologies Laboratory, Institut Pasteur du Maroc, Casablanca, Morocco
| | - Valérie Michel
- Équipe DMic01-Infection, Génotoxicité et Cancer, Département de Microbiologie, Centre National de la Recherche Scientifique (CNRS) Unité Mixte de Recherche (UMR) 6047, Institut Pasteur, Université Paris Cité, 75015, Paris, France
| | - Wafa Badre
- Gastroenterology Department, Ibn Rochd University Hospital Center, Casablanca, Morocco
| | - Eliette Touati
- Équipe DMic01-Infection, Génotoxicité et Cancer, Département de Microbiologie, Centre National de la Recherche Scientifique (CNRS) Unité Mixte de Recherche (UMR) 6047, Institut Pasteur, Université Paris Cité, 75015, Paris, France.
| | - Fatima Maachi
- Helicobacter Pylori and Gastric Pathologies Laboratory, Institut Pasteur du Maroc, Casablanca, Morocco.
| |
Collapse
|
2
|
Srivastava A, Vinod PK. A single-cell network approach to decode metabolic regulation in gynecologic and breast cancers. NPJ Syst Biol Appl 2025; 11:26. [PMID: 40082472 PMCID: PMC11906788 DOI: 10.1038/s41540-025-00506-0] [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: 09/19/2024] [Accepted: 03/03/2025] [Indexed: 03/16/2025] Open
Abstract
Cancer metabolism is characterized by significant heterogeneity, presenting challenges for treatment efficacy and patient outcomes. Understanding this heterogeneity and its regulatory mechanisms at single-cell resolution is crucial for developing personalized therapeutic strategies. In this study, we employed a single-cell network approach to characterize malignant heterogeneity in gynecologic and breast cancers, focusing on the transcriptional regulatory mechanisms driving metabolic alterations. By leveraging single-cell RNA sequencing (scRNA-seq) data, we assessed the metabolic pathway activities and inferred cancer-specific protein-protein interactomes (PPI) and gene regulatory networks (GRNs). We explored the crosstalk between these networks to identify key alterations in metabolic regulation. Clustering cells by metabolic pathways revealed tumor heterogeneity across cancers, highlighting variations in oxidative phosphorylation, glycolysis, cholesterol, fatty acid, hormone, amino acid, and redox metabolism. Our analysis identified metabolic modules associated with these pathways, along with their key transcriptional regulators. These findings provide insights into the complex interplay between metabolic rewiring and transcriptional regulation in gynecologic and breast cancers, paving the way for potential targeted therapeutic strategies in precision oncology. Furthermore, this pipeline for dissecting coregulatory metabolic networks can be broadly applied to decipher metabolic regulation in any disease at single-cell resolution.
Collapse
Affiliation(s)
- Akansha Srivastava
- Centre for Computational Natural Sciences and Bioinformatics, IIIT, Hyderabad, India
| | - P K Vinod
- Centre for Computational Natural Sciences and Bioinformatics, IIIT, Hyderabad, India.
| |
Collapse
|
3
|
Yuasa K, Masubuchi A, Okada T, Shinya M, Inomata Y, Kida H, Shyouji S, Ichikawa H, Takahashi T, Muroi M, Hijikata T. Interferon-Dependent Expression of the Human STAT1 Gene Requires a Distal Regulatory Region Located Approximately 6 kb Upstream for Its Autoregulatory System. Genes Cells 2025; 30:e13188. [PMID: 39722200 DOI: 10.1111/gtc.13188] [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: 11/09/2024] [Revised: 12/12/2024] [Accepted: 12/12/2024] [Indexed: 12/28/2024]
Abstract
We previously suggested that the signal transducer and activator of transcription 1 (STAT1) gene is autoregulated in an interferon (IFN)-dependent manner via a distal regulatory region approximately 5.5-6.2 kb upstream of the murine and human STAT1 promoters (designated 5.5URR). Here, we examined whether this IFN-dependent positive feedback mechanism of the STAT1 gene actually functions in cells. First, we created human embryonic kidney 293 cell mutants lacking the IFN-responsive transcription factor binding sites (IFN-stimulated response element and IFN-gamma-activated sequence) within the 5.5URR and stimulated them with IFN-α/γ. The mutants showed a loss of response to IFN, indicating that the 5.5URR is essential for IFN-induced transcriptional enhancement in STAT1 gene expression. Second, we cloned the full-length 11 kb human STAT1 promoter, including the region upstream of the 5.5URR, from the start codon and linked it to a luciferase gene. Reporter assays showed that IFN-α/γ significantly activated the STAT1 promoter via the 5.5URR. Furthermore, recombinant DNA linking the full-length STAT1 promoter to STAT1 cDNA was introduced into STAT1-deficient cells. In vitro reconstitution experiments showed that IFN-α/γ stimulation increased STAT1 protein levels via the 5.5URR. These results demonstrate that the 5.5URR confers IFN-dependent autoregulation of the STAT1 promoter.
Collapse
Affiliation(s)
- Katsutoshi Yuasa
- Department of Anatomy and Cell Biology, Research Institute of Pharmaceutical Science, Faculty of Pharmacy, Musashino University, Tokyo, Japan
- Research Center for Clinical Pharmacy, Faculty of Pharmacy, Musashino University, Tokyo, Japan
| | - Aimi Masubuchi
- Department of Anatomy and Cell Biology, Research Institute of Pharmaceutical Science, Faculty of Pharmacy, Musashino University, Tokyo, Japan
| | - Tomo Okada
- Department of Anatomy and Cell Biology, Research Institute of Pharmaceutical Science, Faculty of Pharmacy, Musashino University, Tokyo, Japan
| | - Miho Shinya
- Department of Anatomy and Cell Biology, Research Institute of Pharmaceutical Science, Faculty of Pharmacy, Musashino University, Tokyo, Japan
| | - Yui Inomata
- Department of Anatomy and Cell Biology, Research Institute of Pharmaceutical Science, Faculty of Pharmacy, Musashino University, Tokyo, Japan
| | - Honoka Kida
- Department of Anatomy and Cell Biology, Research Institute of Pharmaceutical Science, Faculty of Pharmacy, Musashino University, Tokyo, Japan
| | - Sayoko Shyouji
- Department of Anatomy and Cell Biology, Research Institute of Pharmaceutical Science, Faculty of Pharmacy, Musashino University, Tokyo, Japan
| | - Hirona Ichikawa
- Department of Anatomy and Cell Biology, Research Institute of Pharmaceutical Science, Faculty of Pharmacy, Musashino University, Tokyo, Japan
| | - Tetsuyuki Takahashi
- Department of Anatomy and Cell Biology, Research Institute of Pharmaceutical Science, Faculty of Pharmacy, Musashino University, Tokyo, Japan
| | - Masashi Muroi
- Department of Host Defense, Research Institute of Pharmaceutical Science, Faculty of Pharmacy, Musashino University, Tokyo, Japan
| | - Takao Hijikata
- Department of Anatomy and Cell Biology, Research Institute of Pharmaceutical Science, Faculty of Pharmacy, Musashino University, Tokyo, Japan
| |
Collapse
|
4
|
Kim J, Yu YS, Choi Y, Lee DH, Han S, Kwon J, Noda T, Ikawa M, Kim D, Kim H, Ballabio A, Kim KI, Baek SH. USF2 and TFEB compete in regulating lysosomal and autophagy genes. Nat Commun 2024; 15:8334. [PMID: 39333072 PMCID: PMC11436898 DOI: 10.1038/s41467-024-52600-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Accepted: 09/15/2024] [Indexed: 09/29/2024] Open
Abstract
Autophagy, a highly conserved self-digestion process crucial for cellular homeostasis, is triggered by various environmental signals, including nutrient scarcity. The regulation of lysosomal and autophagy-related processes is pivotal to maintaining cellular homeostasis and basal metabolism. The consequences of disrupting or diminishing lysosomal and autophagy systems have been investigated; however, information on the implications of hyperactivating lysosomal and autophagy genes on homeostasis is limited. Here, we present a mechanism of transcriptional repression involving upstream stimulatory factor 2 (USF2), which inhibits lysosomal and autophagy genes under nutrient-rich conditions. We find that USF2, together with HDAC1, binds to the CLEAR motif within lysosomal genes, thereby diminishing histone H3K27 acetylation, restricting chromatin accessibility, and downregulating lysosomal gene expression. Under starvation, USF2 competes with transcription factor EB (TFEB), a master transcriptional activator of lysosomal and autophagy genes, to bind to target gene promoters in a phosphorylation-dependent manner. The GSK3β-mediated phosphorylation of the USF2 S155 site governs USF2 DNA-binding activity, which is involved in lysosomal gene repression. These findings have potential applications in the treatment of protein aggregation-associated diseases, including α1-antitrypsin deficiency. Notably, USF2 repression is a promising therapeutic strategy for lysosomal and autophagy-related diseases.
Collapse
Affiliation(s)
- Jaebeom Kim
- Creative Research Initiatives Center for Epigenetic Code and Diseases, School of Biological Sciences, Seoul National University, Seoul, South Korea
| | - Young Suk Yu
- Creative Research Initiatives Center for Epigenetic Code and Diseases, School of Biological Sciences, Seoul National University, Seoul, South Korea
| | - Yehwa Choi
- Creative Research Initiatives Center for Epigenetic Code and Diseases, School of Biological Sciences, Seoul National University, Seoul, South Korea
| | - Do Hui Lee
- Creative Research Initiatives Center for Epigenetic Code and Diseases, School of Biological Sciences, Seoul National University, Seoul, South Korea
| | - Soobin Han
- Creative Research Initiatives Center for Epigenetic Code and Diseases, School of Biological Sciences, Seoul National University, Seoul, South Korea
| | - Junhee Kwon
- Department of Biological Sciences, Sookmyung Women's University, Seoul, Republic of Korea
| | - Taichi Noda
- Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
- Institute of Resource Development and Analysis, Kumamoto University, Kumamoto, Japan
- Priority Organization for Innovation and Excellence, Kumamoto University, Kumamoto, Japan
| | - Masahito Ikawa
- Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
- The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Dongha Kim
- Department of Anatomy, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Hyunkyung Kim
- Department of Biochemistry and Molecular Biology, Korea University College of Medicine, Seoul, Republic of Korea
- BK21 Graduate Program, Department of Biomedical Sciences, Korea University College of Medicine, Seoul, Republic of Korea
| | - Andrea Ballabio
- Telethon Institute of Genetics and Medicine (TIGEM), Naples, Italy
- Medical Genetics Unit, Department of Medical and Translational Science, Federico II University, Naples, Italy
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, USA
| | - Keun Il Kim
- Department of Biological Sciences, Sookmyung Women's University, Seoul, Republic of Korea.
| | - Sung Hee Baek
- Creative Research Initiatives Center for Epigenetic Code and Diseases, School of Biological Sciences, Seoul National University, Seoul, South Korea.
| |
Collapse
|
5
|
Nagy G, Bojcsuk D, Tzerpos P, Cseh T, Nagy L. Lineage-determining transcription factor-driven promoters regulate cell type-specific macrophage gene expression. Nucleic Acids Res 2024; 52:4234-4256. [PMID: 38348998 PMCID: PMC11077085 DOI: 10.1093/nar/gkae088] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 01/18/2024] [Accepted: 01/29/2024] [Indexed: 05/09/2024] Open
Abstract
Mammalian promoters consist of multifarious elements, which make them unique and support the selection of the proper transcript variants required under diverse conditions in distinct cell types. However, their direct DNA-transcription factor (TF) interactions are mostly unidentified. Murine bone marrow-derived macrophages (BMDMs) are a widely used model for studying gene expression regulation. Thus, this model serves as a rich source of various next-generation sequencing data sets, including a large number of TF cistromes. By processing and integrating the available cistromic, epigenomic and transcriptomic data from BMDMs, we characterized the macrophage-specific direct DNA-TF interactions, with a particular emphasis on those specific for promoters. Whilst active promoters are enriched for certain types of typically methylatable elements, more than half of them contain non-methylatable and prototypically promoter-distal elements. In addition, circa 14% of promoters-including that of Csf1r-are composed exclusively of 'distal' elements that provide cell type-specific gene regulation by specialized TFs. Similar to CG-rich promoters, these also contain methylatable CG sites that are demethylated in a significant portion and show high polymerase activity. We conclude that this unusual class of promoters regulates cell type-specific gene expression in macrophages, and such a mechanism might exist in other cell types too.
Collapse
Affiliation(s)
- Gergely Nagy
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Dóra Bojcsuk
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Petros Tzerpos
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Tímea Cseh
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - László Nagy
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
- Departments of Medicine and Biological Chemistry, Johns Hopkins University School of Medicine, Institute for Fundamental Biomedical Research, Johns Hopkins All Children's Hospital, St. Petersburg, FL, USA
| |
Collapse
|
6
|
Chen M, Yang Y, Cui J, Qiu L, Zou X, Zeng X. Upstream Stimulating Factor 2 Aggravates Spinal Nerve Ligation-Induced Neuropathic Pain in Mice via Regulating SNHG5/miR-181b-5p. Dev Neurosci 2024; 47:1-11. [PMID: 38471480 DOI: 10.1159/000538178] [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: 06/29/2023] [Accepted: 02/13/2024] [Indexed: 03/14/2024] Open
Abstract
INTRODUCTION Upstream stimulating factor 2 (USF2) belongs to basic Helix-Loop-Helix-Leucine zipper transcription factor family, regulating expression of genes involved in immune response or energy metabolism network. Role of USF2 in neuropathic pain was evaluated. METHODS Mice were intraspinally injected with adenovirus for knockdown of USF2 (Ad-shUSF2) and then subjected to spinal nerve ligation (SNL) to induce neuropathic pain. Distribution and expression of USF2 were detected by western blot and immunofluorescence. Mechanical and thermal pain sensitivity were examined by paw withdrawal thresholds (PWT) and paw withdrawal latency (PWL). Chromatin immunoprecipitation (ChIP) and luciferase activity assays were performed to detect binding ability between USF2 and SNHG5. RESULTS The expression of USF2 was elevated and colocalized with astrocytes and microglia in L5 dorsal root ganglion (DRG) of SNL-induced mice. Injection of Ad-shUSF2 attenuated SNL-induced decrease of PWT and PWL in mice. Knockdown of USF2 increased the level of IL-10 but decreased TNF-α, IL-1β, and IL-6 in SNL-induced mice. Silence of USF2 enhanced protein expression of CD206 while reducing expression of CD16 and CD32 in SNL-induced mice. USF2 binds to promoter of SNHG5 and weakens SNL-induced up-regulation of SNHG5. SNHG5 binds to miR-181b-5p, and miR-181b-5p to interact with CXCL5. CONCLUSION Silence of USF2 ameliorated neuropathic pain, suppressed activation of M1 microglia, and inhibited inflammation in SNL-induced mice through regulation of SNHG5/miR-181b-5p/CXCL5 axis. Therefore, USF2/SNHG5/miR-181b-5p/CXCL5 might be a promising target for neuropathic pain. However, the effect of USF2/SNHG5/miR-181b-5p/CXCL5 on neuropathic pain should also be investigated in further research.
Collapse
Affiliation(s)
- Mi Chen
- Department of Anesthesiology, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Yang Yang
- Department of Anesthesiology, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Jiatian Cui
- Department of Anesthesiology, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Li Qiu
- Department of Anesthesiology, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Xiaohua Zou
- Department of Anesthesiology, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Xianggang Zeng
- Department of Anesthesiology, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| |
Collapse
|
7
|
Chen B, Zhao Y, Xu S, Jiang F, Nie Y, Tang A, Zhou Q. USF2 promotes autophagy and proliferation in chronic lymphocytic leukemia by inhibiting STUB1-induced NFAT5 ubiquitination. Ann Hematol 2024; 103:533-544. [PMID: 37950051 DOI: 10.1007/s00277-023-05522-w] [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: 09/13/2023] [Accepted: 10/25/2023] [Indexed: 11/12/2023]
Abstract
Chronic lymphocytic leukemia (CLL) mainly affects the health of older adults and is difficult to cure. Upstream stimulatory factor 2 (USF2) has been implicated in several diseases and conditions including cancers. However, the effect of USF2 on CLL has not been elucidated. To investigate the effect of USP2 on proliferation and autophagy of CLL, and to explore the underlying mechanism. The mRNA of USF2 and STIP1 homology and U-Box containing protein 1 (STUB1) was analyzed using qRT-PCR. Western blots were used to evaluate the expression level of USF2, LC3II, Beclin-1, P62, STUB1, and NFAT5. The cell proliferation was evaluated using CCK-8 and EdU assays. The cell apoptosis was evaluated using flow cytometry. Indirect fluorescent assay (IFA) was performed to analyze LC3 signal. Nuclear factor of activated T-cells 5 (NFAT5) ubiquitination was detected using immunoprecipitation (IP) assay. The CLL progression was evaluated in xenotransplantation model of nude mice. USF2 was highly expressed in CLL tissues and cell lines. USF2 knockdown suppressed the cell viability and EdU incorporation, while promoting cell apoptosis. Meanwhile, USF2 knockdown reduced the level of LC3II and Beclin-1, but increased P62, illustrating USF2 knockdown inhibiting autophagy. USF2 induced NFAT5 ubiquitination and promoted NFAT5 protein level via repressing STUB1. The downregulation of USF2 weakened CLL progression in xenotransplantation model of nude mice. CLL survival and autophagy was dependent on highly expressed USF2 which promoted the expression and ubiquitination of NFAT5 through inhibiting the transcription of STUB1, which makes USF2 a promising therapeutic candidate for CLL treatment.
Collapse
Affiliation(s)
- Beili Chen
- Department of Hematology, Affiliated Hospital of Guilin Medical University, No. 15, Lequn Road, Xiufeng District, Guilin, 541001, Guangxi, China.
| | - Yanyi Zhao
- Department of Oncology, Affiliated Hospital of Guilin Medical University, Guilin, 541001, Guangxi, China
| | - Shujuan Xu
- Department of Hematology, Affiliated Hospital of Guilin Medical University, No. 15, Lequn Road, Xiufeng District, Guilin, 541001, Guangxi, China
| | - Fang Jiang
- Department of Hematology, Affiliated Hospital of Guilin Medical University, No. 15, Lequn Road, Xiufeng District, Guilin, 541001, Guangxi, China
| | - Yuwei Nie
- Department of Hematology, Affiliated Hospital of Guilin Medical University, No. 15, Lequn Road, Xiufeng District, Guilin, 541001, Guangxi, China
| | - Ailin Tang
- Department of Hematology, Affiliated Hospital of Guilin Medical University, No. 15, Lequn Road, Xiufeng District, Guilin, 541001, Guangxi, China
| | - Qin Zhou
- Department of Hematology, Affiliated Hospital of Guilin Medical University, No. 15, Lequn Road, Xiufeng District, Guilin, 541001, Guangxi, China
| |
Collapse
|
8
|
Zeng YL, Gao F, Zhang C, Ren PP, Ma L, Wang X, Wang R, Kang Y, Li K. USF1 modulates transcription and cellular functions by regulating multiple transcription factors in Huh7 cells. Oncol Lett 2023; 26:532. [PMID: 38020298 PMCID: PMC10655063 DOI: 10.3892/ol.2023.14119] [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: 06/26/2023] [Accepted: 09/28/2023] [Indexed: 12/01/2023] Open
Abstract
Liver cancer, including hepatocellular carcinoma (HCC), is a malignant tumor that has high rates of metastasis and mortality worldwide. Upstream transcription factor 1 (USF1) is a canonical transcription factor (TF) and is associated with the pathogenesis of several cancers, but its biological functions and molecular targets in HCC remain unclear. Huh7 cells that overexpress USF1 were used with whole transcriptome profiling through RNA sequencing and chromatin immunoprecipitation (ChIP) sequencing methods to investigate the downstream targets of USF1. Reverse transcription-quantitative PCR was then used to validate the downstream targets. The results showed that USF1 significantly regulates 350 differentially expressed genes (DEGs). The upregulated DEGs were primarily protein-coding genes enriched in immune and inflammation response pathways, while the downregulated DEGs were mainly coding long non-coding (lnc)RNAs, indicating the regulatory function of USF1. It was also demonstrated that USF1 directly binds to the promoter region of 2,492 genes, which may be involved in the viral progression and cell proliferation pathways. By integrating these two datasets, 16 overlapped genes were detected, including downregulated lncRNA-NEAT1 and upregulated TF-ETV5. The downregulated lncRNA-NEAT1 showed reverse expression pattern and prognosis result compared with that of USF1 in patients with liver cancer, while upregulated TF-ETV5 showed consistent results with USF1. Promoter region motif analysis indicated that ETV5 has more binding motifs and genes than USF1 itself for USF1-regulated DEGs, indicating that USF1 may indirectly modulate gene expression by regulating ETV5 expression in Huh7 cells. The study also validated the direct interaction between USF1 and the promoter of ETV5 using ChIP-qPCR. In summary, the results demonstrated that USF1 binds to the promoter region of thousands of genes and affects a large part of DEGs indirectly. Downstream genes, including lncRNA-NEAT1 and TF-ETV5, may also have potential functions in the regulated network by USF1 and have potential functions in the progression of HCC. The present findings suggested that USF1 and its downstream targets could be potential targets for HCC therapy in the future.
Collapse
Affiliation(s)
- Yan-Li Zeng
- Department of Infectious Diseases, Henan Key Laboratory for Infectious Diseases, Henan Provincial People's Hospital, Zhengzhou, Henan 450003, P.R. China
- Department of Infectious Diseases, Zhengzhou University People's Hospital, Zhengzhou, Henan 450003, P.R. China
- Department of Infectious Diseases, Henan University People's Hospital, Zhengzhou, Henan 450003, P.R. China
| | - Fei Gao
- Department of Infectious Diseases, Henan Key Laboratory for Infectious Diseases, Henan Provincial People's Hospital, Zhengzhou, Henan 450003, P.R. China
| | - Can Zhang
- Department of Infectious Diseases, Henan Key Laboratory for Infectious Diseases, Henan Provincial People's Hospital, Zhengzhou, Henan 450003, P.R. China
| | - Pei-Pei Ren
- Department of Infectious Diseases, Henan Key Laboratory for Infectious Diseases, Henan Provincial People's Hospital, Zhengzhou, Henan 450003, P.R. China
| | - Li Ma
- Department of Infectious Diseases, Henan Key Laboratory for Infectious Diseases, Henan Provincial People's Hospital, Zhengzhou, Henan 450003, P.R. China
| | - Xin Wang
- Department of Infectious Diseases, Henan University, Zhengzhou, Henan 450003, P.R. China
| | - Ruzhen Wang
- Department of Infectious Diseases, Henan Key Laboratory for Infectious Diseases, Henan Provincial People's Hospital, Zhengzhou, Henan 450003, P.R. China
| | - Yi Kang
- Department of Infectious Diseases, Henan Key Laboratory for Infectious Diseases, Henan Provincial People's Hospital, Zhengzhou, Henan 450003, P.R. China
| | - Ke Li
- Department of Infectious Diseases, Henan Key Laboratory for Infectious Diseases, Henan Provincial People's Hospital, Zhengzhou, Henan 450003, P.R. China
| |
Collapse
|
9
|
Liu X, Wang ZZ, Meng S, Zang F, Zhang H, Wang J, Chen YZ. Systematic analysis reveals distinct roles of USF family proteins in various cancer types. Int J Biol Markers 2023; 38:243-252. [PMID: 37846061 DOI: 10.1177/03936155231206135] [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: 10/18/2023]
Abstract
BACKGROUND Upstream stimulatory factors (USFs) are members of the basic helix-loop-helix leucine zipper transcription factor family, including USF1, USF2, and USF3. The first two members have been well studied compared to the third member, USF3, which has received scarce attention in cancer research to date. Despite a recently reported association of its alteration with thyroid carcinoma, its expression has not been previously analyzed. METHODS We comprehensively analyzed differential levels of USFs expression, genomic alteration, DNA methylation, and their prognostic value across different cancer types and the possible correlation with tumor-infiltrating immune cells and drug response by using different bioinformatics tools. RESULTS Our findings established that USFs play an important role in cancers related to the urinary system and justify the necessity for further investigation. We implemented and offer a useful ShinyApp to facilitate researchers' efforts to inquire about any other gene of interest and to perform the analysis of drug response in a user-friendly fashion at http://zzdlab.com:3838/Drugdiscovery/.
Collapse
Affiliation(s)
- Xia Liu
- Department of Lymphoma, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's clinical research center for cancer, Sino-US Center for Lymphoma and Leukemia Research, Tianjin, China
| | - Zhuo-Zhi Wang
- School of Biomedical Engineering, Tianjin Medical University, Tianjin, China
| | - Shuai Meng
- Department of Pharmacy, Key Laboratory of Cancer Prevention and Therapy, Tianjin's clinical research center for cancer, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin, China
| | - Fenglin Zang
- Department of Pathology, Key Laboratory of Cancer Prevention and Therapy, Tianjin's clinical research center for cancer, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin, China
| | - Huilai Zhang
- Department of Lymphoma, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's clinical research center for cancer, Sino-US Center for Lymphoma and Leukemia Research, Tianjin, China
| | - Ju Wang
- School of Biomedical Engineering, Tianjin Medical University, Tianjin, China
| | - Yong-Zi Chen
- Laboratory of Tumor Cell Biology, Key Laboratory of Cancer Prevention and Therapy, Tianjin's clinical research center for cancer, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin, China
| |
Collapse
|
10
|
Fu D, Si Q, Yu C, Han Z, Zang L. USF1-mediated ALKBH5 stabilizes FLII mRNA in an m6A-YTHDF2-dependent manner to repress glycolytic activity in prostate adenocarcinoma. Mol Carcinog 2023; 62:1700-1716. [PMID: 37493109 DOI: 10.1002/mc.23609] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 07/11/2023] [Accepted: 07/12/2023] [Indexed: 07/27/2023]
Abstract
Upstream-stimulating factor 1 (USF1) is a ubiquitously expressed transcription factor implicated in multiple cellular processes, including metabolism and proliferation. This study focused on the function of USF1 in glycolysis and the malignant development of prostate adenocarcinoma (PRAD). Bioinformatics predictions suggested that USF1 is poorly expressed in PRAD. The clinical PRAD samples revealed a low level of USF1, which was correlated with an unfavorable prognosis. Artificial upregulation of USF1 significantly repressed glycolytic activity in PRAD cells and reduced cell growth and metastasis in vitro and in vivo. Potential downstream genes of USF1 were probed by integrated bioinformatics analyses. The chromatin immunoprecipitation and luciferase assays indicated that USF1 bound to the α-ketoglutarate-dependent dioxygenase alkB homolog 5 (ALKBH5) promoter for transcription activation. Flightless I (FLII) was identified as the gene showing the highest degree of correlation with ALKBH5. As an m6A demethylase, ALKBH5 enhanced FLII mRNA stability by inducing m6A demethylation in an m6A-YTH N6-methyladenosine RNA-binding protein F2 (YTHDF2)-dependent manner. Either silencing of ALKBH5 or FLII blocked the role of USF1 in PARD cells and restored glycolysis, cell proliferation, and invasion. This study demonstrates that USF1 activates ALKBH5 to stabilize FLII mRNA in an m6A-YTHDF2-dependent manner, thereby repressing glycolysis processes and the progression of PRAD.
Collapse
Affiliation(s)
- Dewang Fu
- Department of Urology Surgery, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, P.R. China
| | - Qingyue Si
- Department of Urology Surgery, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, P.R. China
| | - Chenxi Yu
- Department of Urology Surgery, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, P.R. China
| | - Zhifu Han
- Department of Urology Surgery, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, P.R. China
| | - Li'e Zang
- Department of Neurology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, P.R. China
| |
Collapse
|
11
|
Huang C, Xia M, Qiao H, Liu Z, Lin Y, Sun H, Yu B, Fang P, Wang J. Tetramerization of upstream stimulating factor USF2 requires the elongated bent leucine zipper of the bHLH-LZ domain. J Biol Chem 2023; 299:105240. [PMID: 37690682 PMCID: PMC10570711 DOI: 10.1016/j.jbc.2023.105240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 09/02/2023] [Accepted: 09/06/2023] [Indexed: 09/12/2023] Open
Abstract
Upstream stimulating factors (USFs), including USF1 and USF2, are key components of the transcription machinery that recruit coactivators and histone-modifying enzymes. Using the classic basic helix-loop-helix leucine zipper (bHLH-LZ) domain, USFs bind the E-box DNA and form tetramers that promote DNA looping for transcription initiation. The structural basis by which USFs tetramerize and bind DNA, however, remains unknown. Here, we report the crystal structure of the complete bHLH-LZ domain of USF2 in complex with E-box DNA. We observed that the leucine zipper (LZ) of USF2 is longer than that of other bHLH-LZ family transcription factors and that the C-terminus of USF2 forms an additional α-helix following the LZ region (denoted as LZ-Ext). We also found the elongated LZ-Ext facilitates compact tetramer formation. In addition to the classic interactions between the basic region and DNA, we show a highly conserved basic residue in the loop region, Lys271, participates in DNA interaction. Together, these findings suggest that USF2 forms a tetramer structure with a bent elongated LZ-Ext region, providing a molecular basis for its role as a key component of the transcription machinery.
Collapse
Affiliation(s)
- Cao Huang
- School of Materials and Chemistry, University of Shanghai for Science and Technology, Shanghai, China
| | - Mingyu Xia
- State Key Laboratory of Chemical Biology, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
| | - Hang Qiao
- State Key Laboratory of Chemical Biology, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
| | - Zaizhou Liu
- State Key Laboratory of Chemical Biology, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
| | - Yuqi Lin
- State Key Laboratory of Chemical Biology, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
| | - Hanyin Sun
- School of Chemistry and Materials Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, China
| | - Biao Yu
- State Key Laboratory of Chemical Biology, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China; School of Chemistry and Materials Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, China
| | - Pengfei Fang
- State Key Laboratory of Chemical Biology, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China; School of Chemistry and Materials Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, China.
| | - Jing Wang
- State Key Laboratory of Chemical Biology, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China; School of Chemistry and Materials Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, China.
| |
Collapse
|
12
|
Pop RT, Pisante A, Nagy D, Martin PCN, Mikheeva L, Hayat A, Ficz G, Zabet NR. Identification of mammalian transcription factors that bind to inaccessible chromatin. Nucleic Acids Res 2023; 51:8480-8495. [PMID: 37486787 PMCID: PMC10484684 DOI: 10.1093/nar/gkad614] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 07/11/2023] [Indexed: 07/26/2023] Open
Abstract
Transcription factors (TFs) are proteins that affect gene expression by binding to regulatory regions of DNA in a sequence specific manner. The binding of TFs to DNA is controlled by many factors, including the DNA sequence, concentration of TF, chromatin accessibility and co-factors. Here, we systematically investigated the binding mechanism of hundreds of TFs by analysing ChIP-seq data with our explainable statistical model, ChIPanalyser. This tool uses as inputs the DNA sequence binding motif; the capacity to distinguish between strong and weak binding sites; the concentration of TF; and chromatin accessibility. We found that approximately one third of TFs are predicted to bind the genome in a DNA accessibility independent fashion, which includes TFs that can open the chromatin, their co-factors and TFs with similar motifs. Our model predicted this to be the case when the TF binds to its strongest binding regions in the genome, and only a small number of TFs have the capacity to bind dense chromatin at their weakest binding regions, such as CTCF, USF2 and CEBPB. Our study demonstrated that the binding of hundreds of human and mouse TFs is predicted by ChIPanalyser with high accuracy and showed that many TFs can bind dense chromatin.
Collapse
Affiliation(s)
- Romana T Pop
- School of Life Sciences, University of Essex, Colchester CO4 3SQ, UK
- Centre for Molecular Medicine Norway (NCMM), Nordic EMBL Partnership, University of Oslo, Oslo, Norway
| | - Alessandra Pisante
- School of Life Sciences, University of Essex, Colchester CO4 3SQ, UK
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London E1 2AT, UK
| | - Dorka Nagy
- School of Life Sciences, University of Essex, Colchester CO4 3SQ, UK
- National Heart and Lung Institute, Imperial College London, London SW3 6LY, UK
| | | | | | - Ateequllah Hayat
- Institute of Medical and Biomedical Education, St George's, University of London, Cranmer Terrace, Tooting SW17 0RE, London
| | - Gabriella Ficz
- Centre for Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, London EC1M 6BQ, UK
| | - Nicolae Radu Zabet
- School of Life Sciences, University of Essex, Colchester CO4 3SQ, UK
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London E1 2AT, UK
| |
Collapse
|
13
|
Kang T, Moore EC, Kopania EEK, King CD, Schilling B, Campisi J, Good JM, Brem RB. A natural variation-based screen in mouse cells reveals USF2 as a regulator of the DNA damage response and cellular senescence. G3 (BETHESDA, MD.) 2023; 13:jkad091. [PMID: 37097016 PMCID: PMC10320765 DOI: 10.1093/g3journal/jkad091] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 03/30/2023] [Accepted: 04/06/2023] [Indexed: 04/26/2023]
Abstract
Cellular senescence is a program of cell cycle arrest, apoptosis resistance, and cytokine release induced by stress exposure in metazoan cells. Landmark studies in laboratory mice have characterized a number of master senescence regulators, including p16INK4a, p21, NF-κB, p53, and C/EBPβ. To discover other molecular players in senescence, we developed a screening approach to harness the evolutionary divergence between mouse species. We found that primary cells from the Mediterranean mouse Mus spretus, when treated with DNA damage to induce senescence, produced less cytokine and had less-active lysosomes than cells from laboratory Mus musculus. We used allele-specific expression profiling to catalog senescence-dependent cis-regulatory variation between the species at thousands of genes. We then tested for correlation between these expression changes and interspecies sequence variants in the binding sites of transcription factors. Among the emergent candidate senescence regulators, we chose a little-studied cell cycle factor, upstream stimulatory factor 2 (USF2), for molecular validation. In acute irradiation experiments, cells lacking USF2 had compromised DNA damage repair and response. Longer-term senescent cultures without USF2 mounted an exaggerated senescence regulatory program-shutting down cell cycle and DNA repair pathways, and turning up cytokine expression, more avidly than wild-type. We interpret these findings under a model of pro-repair, anti-senescence regulatory function by USF2. Our study affords new insights into the mechanisms by which cells commit to senescence, and serves as a validated proof of concept for natural variation-based regulator screens.
Collapse
Affiliation(s)
- Taekyu Kang
- Buck Institute for Research on Aging, Novato, CA 94945, USA
- Department of Plant and Microbial Biology, University of California, Berkeley, CA 94720, USA
| | - Emily C Moore
- Division of Biological Sciences, University of Montana, Missoula, MT 59812, USA
| | - Emily E K Kopania
- Division of Biological Sciences, University of Montana, Missoula, MT 59812, USA
| | | | | | - Judith Campisi
- Buck Institute for Research on Aging, Novato, CA 94945, USA
| | - Jeffrey M Good
- Division of Biological Sciences, University of Montana, Missoula, MT 59812, USA
| | - Rachel B Brem
- Buck Institute for Research on Aging, Novato, CA 94945, USA
- Department of Plant and Microbial Biology, University of California, Berkeley, CA 94720, USA
| |
Collapse
|
14
|
Cho MJ, Lee DG, Lee JW, Hwang B, Yoon SJ, Lee SJ, Park YJ, Park SH, Lee HG, Kim YH, Lee CH, Lee J, Lee NK, Han TS, Cho HS, Moon JH, Lee GS, Bae KH, Hwang GS, Lee SH, Chung SJ, Shim S, Cho J, Oh GT, Kwon YG, Park JG, Min JK. Endothelial PTP4A1 mitigates vascular inflammation via USF1/A20 axis-mediated NF-κB inactivation. Cardiovasc Res 2023; 119:1265-1278. [PMID: 36534975 PMCID: PMC10411943 DOI: 10.1093/cvr/cvac193] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 09/08/2022] [Accepted: 11/08/2022] [Indexed: 12/23/2022] Open
Abstract
AIMS The nuclear factor-κB (NF-κB) signalling pathway plays a critical role in the pathogenesis of multiple vascular diseases. However, in endothelial cells (ECs), the molecular mechanisms responsible for the negative regulation of the NF-κB pathway are poorly understood. In this study, we investigated a novel role for protein tyrosine phosphatase type IVA1 (PTP4A1) in NF-κB signalling in ECs. METHODS AND RESULTS In human tissues, human umbilical artery ECs, and mouse models for loss of function and gain of function of PTP4A1, we conducted histological analysis, immunostaining, laser-captured microdissection assay, lentiviral infection, small interfering RNA transfection, quantitative real-time PCR and reverse transcription-PCR, as well as luciferase reporter gene and chromatin immunoprecipitation assays. Short hairpin RNA-mediated knockdown of PTP4A1 and overexpression of PTP4A1 in ECs indicated that PTP4A1 is critical for inhibiting the expression of cell adhesion molecules (CAMs). PTP4A1 increased the transcriptional activity of upstream stimulatory factor 1 (USF1) by dephosphorylating its S309 residue and subsequently inducing the transcription of tumour necrosis factor-alpha-induced protein 3 (TNFAIP3/A20) and the inhibition of NF-κB activity. Studies on Ptp4a1 knockout or transgenic mice demonstrated that PTP4A1 potently regulates the interleukin 1β-induced expression of CAMs in vivo. In addition, we verified that PTP4A1 deficiency in apolipoprotein E knockout mice exacerbated high-fat high-cholesterol diet-induced atherogenesis with upregulated expression of CAMs. CONCLUSION Our data indicate that PTP4A1 is a novel negative regulator of vascular inflammation by inducing USF1/A20 axis-mediated NF-κB inactivation. Therefore, the expression and/or activation of PTP4A1 in ECs might be useful for the treatment of vascular inflammatory diseases.
Collapse
Affiliation(s)
- Min Ji Cho
- Biotherapeutics Translational Research Centre, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea
| | - Dong Gwang Lee
- Biotherapeutics Translational Research Centre, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea
| | - Jeong Woong Lee
- Biotherapeutics Translational Research Centre, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea
| | - Byungtae Hwang
- Biotherapeutics Translational Research Centre, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea
| | - Sung-Jin Yoon
- Environmental Disease Research Centre, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea
| | - Seon-Jin Lee
- Environmental Disease Research Centre, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea
| | - Young-Jun Park
- Environmental Disease Research Centre, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea
| | - Seung-Ho Park
- Environmental Disease Research Centre, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea
| | - Hee Gu Lee
- Immunotherapy Convergence Research Centre, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea
| | - Yong-Hoon Kim
- Laboratory Animal Resource Centre, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea
| | - Chul-Ho Lee
- Laboratory Animal Resource Centre, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea
| | - Jangwook Lee
- Biotherapeutics Translational Research Centre, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea
| | - Nam-Kyung Lee
- Biotherapeutics Translational Research Centre, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea
| | - Tae-Su Han
- Biotherapeutics Translational Research Centre, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea
| | - Hyun-Soo Cho
- Stem Cell Convergence Research Centre, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea
| | - Jeong Hee Moon
- Disease Target Structure Research Centre, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea
| | - Ga Seul Lee
- Disease Target Structure Research Centre, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea
| | - Kwang-Hee Bae
- Metabolic Regulation Research Centre, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea
| | - Geum-Sook Hwang
- Integrated Metabolomics Research Group, Western Seoul Centre, Korea Basic Science Institute, 150 Bugahyeon-ro, Seodaemun-gu, Seoul 03759, Republic of Korea
| | - Sang-Hak Lee
- Division of Cardiology, Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Sang J Chung
- School of Pharmacy, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon 16419, Republic of Korea
| | - Sungbo Shim
- Department of Biochemistry, Chungbuk National University, 1 Chungdae-ro, Seowon-gu, Cheongju 28644, Republic of Korea
| | - Jaehyung Cho
- Division of Haematology, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Goo Taeg Oh
- Heart-Immune-Brain Network Research Centre, Ewha Womans University, 52 Ewhayeodae-gil, Seodaemun-gu, Seoul 03760, Republic of Korea
| | - Young-Guen Kwon
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Jong-Gil Park
- Biotherapeutics Translational Research Centre, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea
- Department of Bioscience, KRIBB School of Bioscience, Korea University of Science and Technology (UST), 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea
| | - Jeong-Ki Min
- Biotherapeutics Translational Research Centre, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea
- Department of Bioscience, KRIBB School of Bioscience, Korea University of Science and Technology (UST), 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea
| |
Collapse
|
15
|
Choi GH, Cho SH, An HJ, Park HS, Lee JY, Ko EJ, Oh SH, Kim OJ, Kim NK. Association between PAI-1 Polymorphisms and Ischemic Stroke in a South Korean Case-Control Cohort. Int J Mol Sci 2023; 24:8041. [PMID: 37175749 PMCID: PMC10178745 DOI: 10.3390/ijms24098041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Revised: 04/24/2023] [Accepted: 04/26/2023] [Indexed: 05/15/2023] Open
Abstract
Stroke is the second leading cause of death in the world. Approximately 80% of strokes are ischemic in origin. Many risk factors have been linked to stroke, including an increased level of plasminogen activator inhibitor-1 (PAI-1). PAI-1 levels increase and remain elevated in blood during the acute phase of ischemic stroke, which can impair fibrinolytic activity, leading to coronary artery disease and arterial thrombotic disorders. Here, we present a case-control study of 574 stroke patients and 425 controls seen for routine health examination or treatment for nonspecific dizziness, nonorganic headache, or anxiety for positive family history of stroke at the Bundang Medical Center in South Korea. Polymorphisms in PAI-1 were identified by polymerase chain reaction/restriction fragment length polymorphism analysis using genomic DNA. Specifically, three variations (-675 4G>5G, 10692T>C, and 12068G>A) were linked to a higher overall prevalence of stroke as well as a higher prevalence of certain stroke subtypes. Haplotype analyses also revealed combinations of these variations (-844G>A, -675 4G>5G, 43G>A, 9785A>G, 10692T>C, 11053T>G, and 12068G>A) that were significantly associated with a higher prevalence of ischemic stroke. To the best of our knowledge, this is the first strong evidence that polymorphic sites in PAI-1 promoter and 3'-UTR regions are associated with higher ischemic stroke risk. Furthermore, the PAI-1 genotypes and haplotypes identified here have potential as clinical biomarkers of ischemic stroke and could improve the prognosis and future management of stroke patients.
Collapse
Affiliation(s)
- Gun Ho Choi
- Department of Biomedical Science, College of Life Science, CHA University, Seongnam 13488, Republic of Korea
| | - Sung Hwan Cho
- Department of Biomedical Science, College of Life Science, CHA University, Seongnam 13488, Republic of Korea
- College of Medicine, Konyang University, 158 Gwanjeodong-ro, Seo-gu, Daejeon 35365, Republic of Korea
| | - Hui Jeong An
- Department of Biomedical Science, College of Life Science, CHA University, Seongnam 13488, Republic of Korea
- College of Life Science, Gangneung-Wonju National University, 7 Jukheon-gil, Gangneung 25457, Republic of Korea
| | - Han Sung Park
- Department of Biomedical Science, College of Life Science, CHA University, Seongnam 13488, Republic of Korea
| | - Jeong Yong Lee
- Department of Biomedical Science, College of Life Science, CHA University, Seongnam 13488, Republic of Korea
| | - Eun Ju Ko
- Department of Biomedical Science, College of Life Science, CHA University, Seongnam 13488, Republic of Korea
| | - Seung Hun Oh
- Department of Neurology, CHA Bundang Medical Center, School of Medicine, CHA University, Seongnam 13496, Republic of Korea
| | - Ok Joon Kim
- Department of Neurology, CHA Bundang Medical Center, School of Medicine, CHA University, Seongnam 13496, Republic of Korea
| | - Nam Keun Kim
- Department of Biomedical Science, College of Life Science, CHA University, Seongnam 13488, Republic of Korea
| |
Collapse
|
16
|
Cosenza G, Martin P, Garro G, Gallo D, Auzino B, Ciampolini R, Pauciullo A. A novel allelic donkey β-lactogobulin I protein isoform generated by a non-AUG translation initiation codon is associated with a nonsynonymous SNP. J Dairy Sci 2023; 106:4158-4170. [PMID: 37080792 DOI: 10.3168/jds.2022-22598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 12/17/2022] [Indexed: 04/22/2023]
Abstract
β-Lactoglobulin I (β-LG I) is one of the most important whey proteins in donkey milk. However, to our knowledge, there has been no study focusing on the full nucleotide sequences of this gene (BLG I). Current investigation of donkey BLG I gene is very limited with only 2 variants (A and B) characterized so far at the protein level. Recently, a new β-LG I variant, with a significantly higher mass (+1,915 Da) than known variants has been detected. In this study, we report the whole nucleotide sequence of the BLG I gene from 2 donkeys, whose milk samples are characterized by the β-LG I SDS-PAGE band with a normal electrophoretic mobility (18,514.25 Da, β-LG I B1 form) the first, and by the presence of a unique β-LG I band with a higher electrophoretic mobility (20,428.5 Da, β-LG I D form) the latter. A high genetic variability was found all over the 2 sequenced BLG I alleles. In particular, 16 polymorphic sites were found in introns, one in the 5' flanking region, 3 SNPs in the 5' untranslated region and one SNP in the coding region (g.458G > A) located at the 40th nucleotide of exon 2 and responsible for the AA substitutions p.Asp28 > Asn in the mature protein. Two SNPs (g.920-922CAC > TGT and g.1871G/A) were genotyped in 93 donkeys of 2 Italian breeds (60 Ragusana and 33 Amiatina, respectively) and the overall frequencies of g.920-922CAC and g.1871A were 0.3065 and 0.043, respectively. Only the rare allele g.1871A was observed to be associated with the slower migrating β-LG I. Considering this genetic diversity and those found in the database, it was possible to deduce at least 5 different alleles (BLG I A, B, B1, C, D) responsible for 4 potential β-LG I translations. Among these alleles, B1 and D are those characterized in the present research, with the D allele of real novel identification. Haplotype data analysis suggests an evolutionary pathway of donkey BLG I gene and a possible phylogenetic map is proposed. Analyses of mRNA secondary structure showed relevant changes in the structures, as consequence of the g.1871G > A polymorphism, that might be responsible for the recognition of an alternative initiation site providing an additional signal peptide. The extension of 19 AA sequence to the mature protein, corresponding to the canonical signal peptide with an additional alanine residue, is sufficient to provide the observed molecular weight of the slower migrating β-LG I encoded by the BLG I D allele.
Collapse
Affiliation(s)
- G Cosenza
- Department of Agricultural Sciences, University of Naples "Federico II," 80055 Portici (Na), Italy.
| | - P Martin
- Université Paris-Saclay, INRAE, MICALIS Institute, PAPPSO, 78350 Jouy-en-Josas, France
| | - G Garro
- Department of Agricultural Sciences, University of Naples "Federico II," 80055 Portici (Na), Italy
| | - D Gallo
- Department of Agricultural Sciences, University of Naples "Federico II," 80055 Portici (Na), Italy
| | - B Auzino
- Department of Veterinary Science, University of Pisa, 56100, Italy
| | - R Ciampolini
- Department of Veterinary Science, University of Pisa, 56100, Italy.
| | - A Pauciullo
- Department of Agriculture, Forest and Food Sciences, University of Torino, 10095 Grugliasco (TO), Italy
| |
Collapse
|
17
|
Cheng X, Wei Y, Zhang Z, Wang F, He J, Wang R, Xu Y, Keerman M, Zhang S, Zhang Y, Bi J, Yao J, He M. Plasma PFOA and PFOS Levels, DNA Methylation, and Blood Lipid Levels: A Pilot Study. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2022; 56:17039-17051. [PMID: 36374530 DOI: 10.1021/acs.est.2c04107] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Exposure to perfluorooctanoic acid (PFOA) and perfluorooctane sulfonate (PFOS) is associated with blood lipids in adults, but the underlying mechanisms remain unclear. This pilot study aimed to investigate the associations between PFOA or PFOS and epigenome-wide DNA methylation and assess the mediating effect of DNA methylation on the PFOA/PFOS-blood lipid association. We measured plasma PFOA/PFOS and leukocyte DNA methylation in 98 patients enrolled from the hospital between October 2018 and August 2019. The median plasma PFOA/PFOS levels were 0.85 and 2.29 ng/mL. Plasma PFOA and PFOS levels were significantly associated with elevated total cholesterol (TC) and low-density lipoprotein cholesterol (LDL) levels. There were 63/87 CpG positions and 8/11 differentially methylated regions (DMRs) associated with plasma PFOA/PFOS levels, respectively. In addition, 5 CpG positions (annotated to AFF3, CREB5, NRG2, USF2, and intergenic region) and one DMR annotated to IRF6 may mediate the association between plasma PFOA/PFOS and LDL levels (mediated proportion from 7.29 to 46.77%); two CpG positions may mediate the association between plasma PFOA/PFOS and TC levels (annotated to CREB5 and USF2, mediated proportion is around 30%). The data suggest that PFOA/PFOS exposure alters DNA methylation. More importantly, the association of PFOA/PFOS with lipid indicators was partly mediated by DNA methylation changes in lipid metabolism-related genes.
Collapse
Affiliation(s)
- Xu Cheng
- Department of Occupational and Environmental Health and State Key Laboratory of Environmental Health for Incubating, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan 430030, China
| | - Yue Wei
- Department of Occupational and Environmental Health and State Key Laboratory of Environmental Health for Incubating, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan 430030, China
- Department of Occupational Health and Environmental Health, School of Public Health, Guangxi Medical University, Nanning 530021, China
| | - Zefang Zhang
- Department of Occupational and Environmental Health and State Key Laboratory of Environmental Health for Incubating, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan 430030, China
| | - Fei Wang
- Department of Occupational and Environmental Health and State Key Laboratory of Environmental Health for Incubating, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan 430030, China
- Department of Occupational Health and Environmental Health, School of Public Health, Guangxi Medical University, Nanning 530021, China
| | - Jia He
- Department of Occupational and Environmental Health and State Key Laboratory of Environmental Health for Incubating, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan 430030, China
| | - Ruixin Wang
- Department of Occupational and Environmental Health and State Key Laboratory of Environmental Health for Incubating, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan 430030, China
| | - Yali Xu
- Department of Occupational and Environmental Health and State Key Laboratory of Environmental Health for Incubating, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan 430030, China
| | - Mulatibieke Keerman
- Department of Occupational and Environmental Health and State Key Laboratory of Environmental Health for Incubating, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan 430030, China
| | - Shiyang Zhang
- Department of Occupational and Environmental Health and State Key Laboratory of Environmental Health for Incubating, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan 430030, China
| | - Ying Zhang
- Department of Occupational and Environmental Health and State Key Laboratory of Environmental Health for Incubating, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan 430030, China
| | - Jiao Bi
- Department of Occupational and Environmental Health and State Key Laboratory of Environmental Health for Incubating, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan 430030, China
| | - Jinqiu Yao
- Department of Occupational and Environmental Health and State Key Laboratory of Environmental Health for Incubating, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan 430030, China
| | - Meian He
- Department of Occupational and Environmental Health and State Key Laboratory of Environmental Health for Incubating, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan 430030, China
| |
Collapse
|
18
|
False-positive IRESes from Hoxa9 and other genes resulting from errors in mammalian 5' UTR annotations. Proc Natl Acad Sci U S A 2022; 119:e2122170119. [PMID: 36037358 PMCID: PMC9456764 DOI: 10.1073/pnas.2122170119] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Hyperconserved genomic sequences have great promise for understanding core biological processes. It has been recently proposed that scores of hyperconserved 5' untranslated regions (UTRs), also known as transcript leaders (hTLs), encode internal ribosome entry sites (IRESes) that drive cap-independent translation, in part, via interactions with ribosome expansion segments. However, the direct functional significance of such interactions has not yet been definitively demonstrated. We provide evidence that the putative IRESes previously reported in Hox gene hTLs are rarely included in transcript leaders. Instead, these regions function independently as transcriptional promoters. In addition, we find the proposed RNA structure of the putative Hoxa9 IRES is not conserved. Instead, sequences previously shown to be essential for putative IRES activity encode a hyperconserved transcription factor binding site (E-box) that contributes to its promoter activity and is bound by several transcription factors, including USF1 and USF2. Similar E-box sequences enhance the promoter activities of other putative Hoxa gene IRESes. Moreover, we provide evidence that the vast majority of hTLs with putative IRES activity overlap transcriptional promoters, enhancers, and 3' splice sites that are most likely responsible for their reported IRES activities. These results argue strongly against recently reported widespread IRES-like activities from hTLs and contradict proposed interactions between ribosomal expansion segment ES9S and putative IRESes. Furthermore, our work underscores the importance of accurate transcript annotations, controls in bicistronic reporter assays, and the power of synthesizing publicly available data from multiple sources.
Collapse
|
19
|
Wang J, Li S, Li X, Liu J, Yang J, Li Y, Li W, Yang Y, Li J, Chen R, Li K, Huang D, Liu Y, Lv L, Li M, Xiao X, Luo XJ. Functional variant rs2270363 on 16p13.3 confers schizophrenia risk by regulating NMRAL1. Brain 2022; 145:2569-2585. [PMID: 35094059 PMCID: PMC9612800 DOI: 10.1093/brain/awac020] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Revised: 11/17/2021] [Accepted: 12/20/2021] [Indexed: 12/28/2023] Open
Abstract
Recent genome-wide association studies have reported multiple schizophrenia risk loci, yet the functional variants and their roles in schizophrenia remain to be characterized. Here we identify a functional single nucleotide polymorphism (rs2270363: G>A) at the schizophrenia risk locus 16p13.3. rs2270363 lies in the E-box element of the promoter of NMRAL1 and disrupts binding of the basic helix-loop-helix leucine zipper family proteins, including USF1, MAX and MXI1. We validated the regulatory effects of rs2270363 using reporter gene assays and electrophoretic mobility shift assay. Besides, expression quantitative trait loci analysis showed that the risk allele (A) of rs2270363 was significantly associated with elevated NMRAL1 expression in the human brain. Transcription factors knockdown and CRISPR-Cas9-mediated editing further confirmed the regulatory effects of the genomic region containing rs2270363 on NMRAL1. Intriguingly, NMRAL1 was significantly downregulated in the brain of schizophrenia patients compared with healthy subjects, and knockdown of Nmral1 expression affected proliferation and differentiation of mouse neural stem cells, as well as genes and pathways associated with brain development and synaptic transmission. Of note, Nmral1 knockdown resulted in significant decrease of dendritic spine density, revealing the potential pathophysiological mechanisms of NMRAL1 in schizophrenia. Finally, we independently confirmed the association between rs2270363 and schizophrenia in the Chinese population and found that the risk allele of rs2270363 was the same in European and Chinese populations. These lines of evidence suggest that rs2270363 may confer schizophrenia risk by regulating NMRAL1, a gene whose expression dysregulation might be involved in the pathogenesis of schizophrenia by affecting neurodevelopment and synaptic plasticity.
Collapse
Affiliation(s)
- Junyang Wang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan 650204, China
| | - Shiwu Li
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan 650204, China
| | - Xiaoyan Li
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan 650204, China
| | - Jiewei Liu
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China
| | - Jinfeng Yang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan 650204, China
| | - Yifan Li
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan 650204, China
| | - Wenqiang Li
- Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan 453002, China
- Henan Key Lab of Biological Psychiatry, International Joint Research Laboratory for Psychiatry and Neuroscience of Henan, Xinxiang Medical University, Xinxiang, Henan 453002, China
| | - Yongfeng Yang
- Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan 453002, China
- Henan Key Lab of Biological Psychiatry, International Joint Research Laboratory for Psychiatry and Neuroscience of Henan, Xinxiang Medical University, Xinxiang, Henan 453002, China
| | - Jiao Li
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan 650204, China
| | - Rui Chen
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan 650204, China
| | - Kaiqin Li
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China
| | - Di Huang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China
| | - Yixing Liu
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan 650204, China
| | - Luxian Lv
- Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan 453002, China
- Henan Key Lab of Biological Psychiatry, International Joint Research Laboratory for Psychiatry and Neuroscience of Henan, Xinxiang Medical University, Xinxiang, Henan 453002, China
| | - Ming Li
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China
| | - Xiao Xiao
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China
| | - Xiong Jian Luo
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan 650204, China
- Zhongda Hospital, School of Life Sciences and Technology, Advanced Institute for Life and Health, Southeast University, Nanjing, Jiangsu 210096, China
- Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, Yunnan 650223, China
| |
Collapse
|
20
|
Zheng PF, Chen LZ, Pan HW, Liu P, Zheng ZF. Effects of USF1 SNPs and SNP–Environment Interactions on Serum Lipid Profiles and the Risk of Early-Onset Coronary Artery Disease in the Chinese Population. Front Cardiovasc Med 2022; 9:882728. [PMID: 35783856 PMCID: PMC9240353 DOI: 10.3389/fcvm.2022.882728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 05/16/2022] [Indexed: 11/13/2022] Open
Abstract
BackgroundUpstream transcription factor 1 (USF1) single-nucleotide polymorphisms (SNPs) are significantly associated with serum lipid levels in several different ethnic groups or populations, but their association with lipid levels and the risk of early-onset coronary artery disease (EOCAD) has not been reported in Han populations of southern China.MethodsSix USF1 SNPs (rs3737787, rs2774276, rs2516839, rs2516838, rs1556259, and rs2516837) were genotyped by next-generation sequencing (NGS) techniques in 686 control subjects and 728 patients with EOCAD.ResultsThe genotypic and allelic frequencies of the USF1 rs3737787 SNP were significantly different between the control and EOCAD groups. The subgroup analysis identified that the rs3737787T allele was related to a decreased risk of EOCAD, whereas the rs3737787C–rs2774276G–rs2516839A and rs3737787C–rs2774276G–rs2516839G haplotypes were related to an increased risk of EOCAD in men, and the rs3737787C–rs2774276G–rs2516839A and rs3737787T–rs2774276C–rs2516839A haplotypes were correlated with an increased risk of EOCAD in women (p < 0.05–0.01). Male rs3737787T allele carriers had lower low-density lipoprotein cholesterol (LDL-C), total cholesterol (TC), and triglyceride (TG) concentrations than the rs3737787T allele non-carriers (p < 0.01). The interactions of rs3737787 with alcohol consumption and rs2516839 with smoking affected serum TC and LDL-C levels in men, whereas the interaction of rs3737787 with alcohol consumption affected serum high-density lipoprotein cholesterol (HDL-C) levels and the rs2516839-smoking interaction affected serum TC levels in women (pI < 0.001). The expression levels of the USF1 mRNA, interleukin 1β (IL-1β), tumor necrosis factor-α (TNF-α), and interleukin 6 (IL-6) were significantly lower in controls than in patients with EOCAD, and rs3737787T allele carriers displayed lower IL-1β, TNF-α, IL-6, and USF1 mRNA expression levels than the rs3737787T allele non-carriers. In addition, IL-1β, TNF-α, and IL-6 expression levels were significantly positively correlated with USF1 mRNA levels (p < 0.01).ConclusionSex-specific correlations were identified between the USF1 rs3737787T allele with blood lipid levels and the risk of EOCAD. The USF1 rs3737787T allele affects the risk of EOCAD by modulating serum lipid levels and the expression of inflammatory factors, including IL-1β, TNF-α, and IL-6.
Collapse
Affiliation(s)
- Peng-Fei Zheng
- Cardiology Department, Hunan Provincial People's Hospital, Changsha, China
- Clinical Research Center for Heart Failure in Hunan Province, Changsha, China
- Institute of Cardiovascular Epidemiology, Hunan Provincial People's Hospital, Changsha, China
| | - Lu-Zhu Chen
- Department of Cardiology, The Central Hospital of ShaoYang, Shaoyang, China
| | - Hong-Wei Pan
- Cardiology Department, Hunan Provincial People's Hospital, Changsha, China
- Clinical Research Center for Heart Failure in Hunan Province, Changsha, China
- Institute of Cardiovascular Epidemiology, Hunan Provincial People's Hospital, Changsha, China
| | - Peng Liu
- Department of Cardiology, The Central Hospital of ShaoYang, Shaoyang, China
- *Correspondence: Peng Liu
| | - Zhao-Fen Zheng
- Cardiology Department, Hunan Provincial People's Hospital, Changsha, China
- Clinical Research Center for Heart Failure in Hunan Province, Changsha, China
- Institute of Cardiovascular Epidemiology, Hunan Provincial People's Hospital, Changsha, China
- Zhao-Fen Zheng
| |
Collapse
|
21
|
Lu S, Louphrasitthiphol P, Goradia N, Lambert JP, Schmidt J, Chauhan J, Rughani MG, Larue L, Wilmanns M, Goding CR. TBX2 controls a proproliferative gene expression program in melanoma. Genes Dev 2021; 35:1657-1677. [PMID: 34819350 PMCID: PMC8653791 DOI: 10.1101/gad.348746.121] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Accepted: 10/22/2021] [Indexed: 12/20/2022]
Abstract
Senescence shapes embryonic development, plays a key role in aging, and is a critical barrier to cancer initiation, yet how senescence is regulated remains incompletely understood. TBX2 is an antisenescence T-box family transcription repressor implicated in embryonic development and cancer. However, the repertoire of TBX2 target genes, its cooperating partners, and how TBX2 promotes proliferation and senescence bypass are poorly understood. Here, using melanoma as a model, we show that TBX2 lies downstream from PI3K signaling and that TBX2 binds and is required for expression of E2F1, a key antisenescence cell cycle regulator. Remarkably, TBX2 binding in vivo is associated with CACGTG E-boxes, present in genes down-regulated by TBX2 depletion, more frequently than the consensus T-element DNA binding motif that is restricted to Tbx2 repressed genes. TBX2 is revealed to interact with a wide range of transcription factors and cofactors, including key components of the BCOR/PRC1.1 complex that are recruited by TBX2 to the E2F1 locus. Our results provide key insights into how PI3K signaling modulates TBX2 function in cancer to drive proliferation.
Collapse
Affiliation(s)
- Sizhu Lu
- Ludwig Institute for Cancer Research, Nuffield Department of Clinical Medicine, University of Oxford, Headington, Oxford OX3 7DQ, United Kingdom
| | - Pakavarin Louphrasitthiphol
- Ludwig Institute for Cancer Research, Nuffield Department of Clinical Medicine, University of Oxford, Headington, Oxford OX3 7DQ, United Kingdom.,Department of Surgery, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Nishit Goradia
- European Molecular Biology Laboratory, Hamburg Unit, 22607 Hamburg, Germany
| | - Jean-Philippe Lambert
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario M5G 1X5, Canada.,Department of Molecular Medicine and Cancer Research Centre, Université Laval, Québec City, Québec G1R 3S3, Canada; CHU de Québec Research Center, Centre Hospitalier de l'Université Laval, Québec City, Québec G1V 4G2, Canada
| | - Johannes Schmidt
- Ludwig Institute for Cancer Research, Nuffield Department of Clinical Medicine, University of Oxford, Headington, Oxford OX3 7DQ, United Kingdom
| | - Jagat Chauhan
- Ludwig Institute for Cancer Research, Nuffield Department of Clinical Medicine, University of Oxford, Headington, Oxford OX3 7DQ, United Kingdom
| | - Milap G Rughani
- Ludwig Institute for Cancer Research, Nuffield Department of Clinical Medicine, University of Oxford, Headington, Oxford OX3 7DQ, United Kingdom
| | - Lionel Larue
- Institut Curie, PSL Research University, U1021, Institut National de la Santé et de la Recherche Médicale, Normal and Pathological Development of Melanocytes, 91405 Orsay Cedex, France.,Université Paris-Sud, Université Paris-Saclay, UMR 3347 Centre National de la Recherche Scientifique, 91405 Orsay Cedex, France.,Equipe Labellisée Ligue Contre le Cancer, 91405 Orsay Cedex, France
| | - Matthias Wilmanns
- European Molecular Biology Laboratory, Hamburg Unit, 22607 Hamburg, Germany.,University Hamburg Clinical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Colin R Goding
- Ludwig Institute for Cancer Research, Nuffield Department of Clinical Medicine, University of Oxford, Headington, Oxford OX3 7DQ, United Kingdom
| |
Collapse
|
22
|
Zeng HB, Dong LQ, Huang YL, Xu C, Zhao XH, Wu LG. USF2 reduces BMP3 expression via transcriptional activation of miR-34a, thus promoting osteogenic differentiation of BMSCs. J Bone Miner Metab 2021; 39:997-1008. [PMID: 34350522 DOI: 10.1007/s00774-021-01254-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 07/18/2021] [Indexed: 11/26/2022]
Abstract
INTRODUCTION Osteoporosis is the most susceptible disease for people over 60. The main cause of osteoporosis is the decreased osteogenic differentiation of mesenchymal stem cells (MSCs). Here we showed that upstream stimulatory factor 2 (USF2)/microRNA-34a (miR-34a)/bone morphogenetic protein 3 (BMP3) axis regulated osteogenic differentiation of BMSCs. MATERIALS AND METHODS USF2 and miR-34a expression were examined using qPCR. Protein levels of BMP3 and osteogenic markers expression were evaluated using both western blot and qPCR. Activity of ALP was determined by ALP assay kit. Mineralization capacity of hBMSCs was assessed using ARS. Besides, CHIP assay was employed to verify whether USF2 could bind to miR-34a promoter. Finally, RIP assay and dual-luciferase reporter assay were employed to verify whether miR-34a directly bound to BMP3. RESULTS Our results suggested that miR-34a was upregulated during osteogenic differentiation of BMSCs, and miR-34a overexpression could enhance osteogenic differentiation of BMSCs. USF2 could positively regulate miR-34a expression by interacting with its promoter. USF2 overexpression enhanced osteogenic differentiation of BMSCs, while miR-34a inhibition reversed the effect. Besides, BMP3 was the target of miR-34a. MiR-34a overexpression enhanced osteogenic differentiation of BMSCs, which was abolished by BMP3 overexpression. CONCLUSION Taken together, USF2 enhanced osteogenic differentiation of BMSCs via downregulating BMP3 by interacting with miR-34a promoter.
Collapse
Affiliation(s)
- Han-Bing Zeng
- Department of Orthopaedics, The Second Affiliated Hospital of Zhejiang Chinese Medical University, No.318, Chaowang Road, Gongshu District, Hangzhou, 310005, Zhejiang Province, China
| | - Li-Qiang Dong
- Department of Orthopaedics, The Second Affiliated Hospital of Zhejiang Chinese Medical University, No.318, Chaowang Road, Gongshu District, Hangzhou, 310005, Zhejiang Province, China
| | - Yu-Liang Huang
- Department of Orthopaedics, The Second Affiliated Hospital of Zhejiang Chinese Medical University, No.318, Chaowang Road, Gongshu District, Hangzhou, 310005, Zhejiang Province, China
| | - Chao Xu
- Department of Orthopaedics, The Second Affiliated Hospital of Zhejiang Chinese Medical University, No.318, Chaowang Road, Gongshu District, Hangzhou, 310005, Zhejiang Province, China
| | - Xu-Hui Zhao
- Department of Orthopaedics, The Second Affiliated Hospital of Zhejiang Chinese Medical University, No.318, Chaowang Road, Gongshu District, Hangzhou, 310005, Zhejiang Province, China
| | - Lian-Guo Wu
- Department of Orthopaedics, The Second Affiliated Hospital of Zhejiang Chinese Medical University, No.318, Chaowang Road, Gongshu District, Hangzhou, 310005, Zhejiang Province, China.
| |
Collapse
|
23
|
Gupta S, Ahmed MM. Targeting radiation-induced upstream stimulatory factor-1 by histone deacetylase inhibitors to reverse radioresistance in prostate cancer. Cancer Rep (Hoboken) 2021; 5:e1553. [PMID: 34533293 PMCID: PMC9780427 DOI: 10.1002/cnr2.1553] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 08/11/2021] [Accepted: 08/23/2021] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND Ionizing radiation (IR) is a standard modality for the management of solid tumors. Apart from its killing effects, IR can induce pro-survival factors leading to radioresistance of cancer. Mechanistic understanding of radiation resistance is warranted to overcome the pro-survival effects of IR. AIM The aim of this study was to investigate the role of upstream stimulatory factor-1 (USF-1) in the induction of radioresistance in prostate cancer and its targeting by histone deacetylase (HDAC) inhibitors to reverse resistance. METHODS AND RESULTS This study reports here that USF-1 is a marker for radioresistance in PC-3 cells. Using protein-DNA array analysis, it was documented that DNA binding activity of USF-1 was elevated following IR in PC-3 cells. Novel HDAC inhibitors downregulated USF-1 binding either alone or in combination with IR. A 5 Gy dose of IR induced the expression of target genes of USF-1 (human telomerase reverse transcriptase [hTERT], IGF2R, CyclinB1, and Cdk1), however, HDAC inhibitors alone or in combination with IR reduced their expression as measured by real time RT PCR analysis. Furthermore, immunofluorescence analysis revealed that while USF-1 localized primarily in the nucleus following IR, it localized in the cytoplasm when treated with HDAC inhibitors/combination. Maximum effects of modulation of USF-1 expression (overexpression or suppression) were observed on hTERT activity as determined by dual-luciferase reporter assay. To further confirm the role of USF-1 in radioresistance, cell growth was analyzed using the real-time cell electronic sensing (RT-CES) system. This study found that USF-1-transfected cells proliferated faster than the vector-transfected cells with or without treatments with HDAC inhibitors/IR/combination. Colony forming assay also confirmed that USF-1 overexpression led to increased survival following IR. Importantly, colony-forming assay demonstrated that HDAC inhibitors reversed the radioresistance in both PC-3 and DU-145 cells. CONCLUSION These studies demonstrate that HDAC inhibitors reverse the radioresistance in prostate cancer through down-modulation of USF-1-mediated transactivation of target genes involved in cell proliferation and cell cycle.
Collapse
Affiliation(s)
- Seema Gupta
- Department of Radiation OncologyUniversity of MiamiMiamiFloridaUSA,Present address:
The Loop Immuno‐Oncology Laboratory, Lombardi Comprehensive Cancer CenterGeorgetown University Medical CenterWashington, DCUSA
| | - Mansoor M. Ahmed
- Department of Radiation OncologyUniversity of MiamiMiamiFloridaUSA,Present address:
Radiation Research Program (RRP), Division of Cancer Treatment and Diagnosis (DCTD)National Cancer Institute/National Institutes of HealthRockvilleMarylandUSA
| |
Collapse
|
24
|
Ding Y, Jiang Y, Im ST, Myung S, Kim HS, Lee SH. Diphlorethohydroxycarmalol inhibits melanogenesis via protein kinase A/cAMP response element-binding protein and extracellular signal-regulated kinase-mediated microphthalmia-associated transcription factor downregulation in α-melanocyte stimulating hormone-stimulated B16F10 melanoma cells and zebrafish. Cell Biochem Funct 2021; 39:546-554. [PMID: 33474761 DOI: 10.1002/cbf.3620] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 12/14/2020] [Accepted: 01/05/2021] [Indexed: 11/08/2022]
Abstract
Diphlorethohydroxycarmalol (DPHC) is a marine polyphenolic compound derived from brown alga Ishige okamurae. A previously study has suggested that DPHC possesses strong mushroom tyrosinase inhibitory activity. However, the anti-melanogenesis effect of DPHC has not been reported at cellular level. The objective of the present study was to clarify the melanogenesis inhibitory effect of DPHC and its molecular mechanisms in murine melanoma cells (B16F10) and zebrafish model. DPHC significantly inhibited tyrosinase activity and melanin content dose-dependently in α-melanocyte stimulating hormone (α-MSH)-stimulated B16F10 cells. This polyphenolic compound also suppressed the expression of phosphorylation of cAMP response element-binding protein (CREB) by attenuating phosphorylation of cAMP-dependent protein kinase A, resulting in decreased MITF expression levels. Furthermore, DPHC downregulated MITF protein expression levels by promoting the phosphorylation of extracellular signal-regulated kinase. It also inhibited tyrosinase, tyrosinase-related protein 1 (TRP-1), and TRP-2 in α-MSH stimulated B16F10 cells. In in vivo studies using zebrafish, DPHC also markedly inhibited melanin synthesis in a dose-dependent manner. These results demonstrate that DPHC can effectively inhibit melanogenesis in melanoma cells in vitro and in zebrafish in vivo, suggesting that DPHC could be applied in fields of pharmaceutical and cosmeceuticals as a skin-whitening agent. Significance of study: The present study showed for the first time that DPHC could inhibit a-MSH-stimulated melanogenesis via PKA/CREB and ERK pathway in melanoma cells. It also could inhibit pigmentation in vivo in a zebrafish model. This evidence suggests that DPHC has potential as a skin whitening agent. Taken together, DPHC could be considered as a novel anti-melanogenic agent to be applied in cosmetic, food, and medical industry.
Collapse
Affiliation(s)
- Yuling Ding
- Department of Biopharmaceuticals, School of Pharmaceutical Sciences, Changchun University of Chinese Medicine, Chanchun, China
| | - Yunfei Jiang
- Department of Marine Life Science, Jeju National University, Jeju, Republic of Korea
| | - Seung Tae Im
- Department of Medical Science, Soonchunhyang University, Asan, Republic of Korea
| | - Seungwon Myung
- Department of Medical Science, Soonchunhyang University, Asan, Republic of Korea
| | - Hyun-Soo Kim
- Department of Applied Research, National Marine Biodiversity Institute of Korea, Seochun, Republic of Korea
| | - Seung-Hong Lee
- Department of Medical Science, Soonchunhyang University, Asan, Republic of Korea
- Department of Pharmaceutical Engineering, Soonchunhyang University, Asan, Republic of Korea
| |
Collapse
|
25
|
Li J, Zhang S, Shen C, Niu Z, Yang H, Zhang K, Liu Z, Wang Y, Lan X. Indel mutations within the bovine HSD17B3 gene are significantly associated with ovary morphological traits and mature follicle number. J Steroid Biochem Mol Biol 2021; 209:105833. [PMID: 33524543 DOI: 10.1016/j.jsbmb.2021.105833] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 01/17/2021] [Accepted: 01/24/2021] [Indexed: 12/24/2022]
Abstract
Given the intensive selection for increased milk production, it is imperative that the problem of declining fertility in dairy cows be reversed. In female mammals their reproductive traits and functioning is controlled by a finely tuned process balancing estrogens and androgens, in which androgens (e.g., testosterone) as a precursor substance can participate in estrogen synthesis by activating 17β-hydroxysteroid dehydrogenase (17betaHSD). Being a key catalyst for testosterone synthesis, we hypothesized HSD17B3 gene is involved in the ovary's development and thereby capable of influencing cows' fecundity. Herein, to investigate the relationship between polymorphisms of the HSD17B3 gene and cow fertility, we characterized three insertion/deletion (indels) polymorphisms of this gene in 1110 healthy bovine ovaries. Their respective minimum allelic frequency (MAF) ranged from 0.180 to 0.482. For the ovary morphological traits, correlations revealed that both P1-D15-bp and P4-D19-bp demonstrated significant associations with ovarian height (P = 0.007 and 0.004, respectively), while P5-I5-bp was found to be significantly associated with the ovarian weight (P = 0.024). For ovarian volume, a significant correlation was uncovered between it and both polymorphisms of P4-D19-bp (P = 0.036) and P5-I5-bp (P = 0.045). Cows with either the DD genotype of P4-D19-bp or P5-I5-bp tended to have greater ovarian volume, a result consistent with their relationship to ovarian weight (P5-I5-bp) or height (P4-D19-bp). For the mature follicle traits, polymorphisms of P4-D19-bp were found significantly associated with the number of mature follicles (P = 0.045). Furthermore, expression levels of HSD17B3 differed significantly between the maximal and minimum groups of ovarian weight or volume, and the transcription factors GATA-1 and USF were predicted to bind P1-D15-bp and P4-D19-bp, respectively. This suggested the detected intron mutations could affect HSD17B3's transcription by regulating the binding of transcription factors, thereby affecting ovarian weight and other reproductive traits. As a potential effective molecular marker loci significantly related to traits of ovary and follicle, these three indels could be used in practical molecular marker-assisted selection (MAS) breeding programs, to optimize female fertility and enhance economic efficiency in the dairy cow industry.
Collapse
Affiliation(s)
- Jie Li
- Animal Genome and Gene Function Laboratory, College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, PR China.
| | - Shaoli Zhang
- Animal Genome and Gene Function Laboratory, College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, PR China.
| | - Chenglong Shen
- College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, PR China.
| | - Zhihan Niu
- College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, PR China.
| | - Han Yang
- Animal Genome and Gene Function Laboratory, College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, PR China.
| | - Kaijuan Zhang
- College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, PR China.
| | - Zhengqing Liu
- College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, PR China.
| | - Yongsheng Wang
- College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, PR China.
| | - Xianyong Lan
- Animal Genome and Gene Function Laboratory, College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, PR China.
| |
Collapse
|
26
|
MacBeth M, Joetham A, Gelfand EW, Schedel M. Plasticity of Naturally Occurring Regulatory T Cells in Allergic Airway Disease Is Modulated by the Transcriptional Activity of Il-6. Int J Mol Sci 2021; 22:ijms22094582. [PMID: 33925531 PMCID: PMC8123826 DOI: 10.3390/ijms22094582] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 04/26/2021] [Accepted: 04/26/2021] [Indexed: 12/27/2022] Open
Abstract
The impact of naturally occurring regulatory T cells (nTregs) on the suppression or induction of lung allergic responses in mice depends on the nuclear environment and the production of the pro-inflammatory cytokine interleukin 6 (IL-6). These activities were shown to be different in nTregs derived from wild-type (WT) and CD8-deficient mice (CD8−/−), with increased IL-6 levels in nTregs from CD8−/− mice in comparison to WT nTregs. Thus, identification of the molecular mechanisms regulating IL-6 production is critical to understanding the phenotypic plasticity of nTregs. Electrophoretic mobility shift assays (EMSA) were performed to determine transcription factor binding to four Il-6 promoter loci using nuclear extracts from nTregs of WT and CD8−/− mice. Increased transcription factor binding for each of the Il-6 loci was identified in CD8−/− compared to WT nTregs. The impact of transcription factor binding and a novel short tandem repeat (STR) on Il-6 promoter activity was analyzed by luciferase reporter assays. The Il-6 promoter regions closer to the transcription start site (TSS) were more relevant to the regulation of Il-6 depending on NF-κB, c-Fos, and SP and USF family members. Two Il-6 promoter loci were most critical for the inducibility by lipopolysaccharide (LPS) and tumor necrosis factor α (TNFα). A novel STR of variable length in the Il-6 promoter was identified with diverging prevalence in nTregs from WT or CD8−/− mice. The predominant GT repeat in CD8−/− nTregs revealed the highest luciferase activity. These novel regulatory mechanisms controlling the transcriptional regulation of the Il-6 promoter are proposed to contribute to nTregs plasticity and may be central to disease pathogenesis.
Collapse
Affiliation(s)
- Morgan MacBeth
- Division of Allergy and Immunology and Cell Biology, Department of Pediatrics, National Jewish Health, Denver, CO 80206, USA; (M.M.); (A.J.); (E.W.G.)
- Department of Medical Oncology, University of Colorado, Denver, CO 80206, USA
| | - Anthony Joetham
- Division of Allergy and Immunology and Cell Biology, Department of Pediatrics, National Jewish Health, Denver, CO 80206, USA; (M.M.); (A.J.); (E.W.G.)
| | - Erwin W. Gelfand
- Division of Allergy and Immunology and Cell Biology, Department of Pediatrics, National Jewish Health, Denver, CO 80206, USA; (M.M.); (A.J.); (E.W.G.)
| | - Michaela Schedel
- Division of Allergy and Immunology and Cell Biology, Department of Pediatrics, National Jewish Health, Denver, CO 80206, USA; (M.M.); (A.J.); (E.W.G.)
- Department of Pulmonary Medicine, University Medical Center Essen-Ruhrlandklinik, 45239 Essen, Germany
- University Hospital Essen, University Duisburg-Essen, 45147 Essen, Germany
- Correspondence: ; Tel.: +49-201-723-82545
| |
Collapse
|
27
|
Ko SC, Lee SH. Protocatechuic Aldehyde Inhibits α-MSH-Induced Melanogenesis in B16F10 Melanoma Cells via PKA/CREB-Associated MITF Downregulation. Int J Mol Sci 2021; 22:ijms22083861. [PMID: 33917915 PMCID: PMC8068260 DOI: 10.3390/ijms22083861] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 04/06/2021] [Accepted: 04/08/2021] [Indexed: 11/16/2022] Open
Abstract
Protocatechuic aldehyde (PA) is a naturally occurring phenolic compound that is a potent inhibitor of mushroom tyrosinase. However, the molecular mechanisms of the anti-melanogenesis activity of PA have not yet been reported. The aim of the current study was to clarify the melanogenesis inhibitory effects of PA and its molecular mechanisms in murine melanoma cells (B16F10). We first predicted the 3D structure of tyrosinase and used a molecular docking algorithm to simulate binding between tyrosinase and PA. These molecular modeling studies calculated a binding energy of -527.42 kcal/mol and indicated that PA interacts with Cu400 and 401, Val283, and His263. Furthermore, PA significantly decreased α-MSH-induced intracellular tyrosinase activity and melanin content in a dose-dependent manner. PA also inhibited key melanogenic proteins such as tyrosinase, tyrosinase-related protein 1 (TRP-1), and TRP-2 in α-MSH-stimulated B16F10 cells. In addition, PA decreased MITF expression levels by inhibiting phosphorylation of cAMP response element-binding protein (CREB) and cAMP-dependent protein kinase A (PKA). These results demonstrate that PA can effectively suppress melanin synthesis in melanoma cells. Taken together, our results show that PA could serve as a potential inhibitor of melanogenesis, and hence could be explored as a possible skin-lightening agent.
Collapse
Affiliation(s)
- Seok-Chun Ko
- Department of Genetic Resources, National Marine Biodiversity Institute of Korea, Seocheon 33662, Korea;
| | - Seung-Hong Lee
- Department of Pharmaceutical Engineering and Medical Science, Soonchunhyang University, Asan 31538, Korea
- Correspondence: ; Tel.: +82-41-530-4980; Fax: +82-41-530-3085
| |
Collapse
|
28
|
Abstract
Evolutionary processes, including mutation, migration and natural selection, have influenced the prevalence and distribution of various disorders in humans. However, despite a few well-known examples, such as the APOL1 variants - which have undergone positive genetic selection for their ability to confer resistance to Trypanosoma brucei infection but confer a higher risk of chronic kidney disease - little is known about the effects of evolutionary processes that have shaped genetic variation on kidney disease. An understanding of basic concepts in evolutionary genetics provides an opportunity to consider how findings from ancient and archaic genomes could inform our knowledge of evolution and provide insights into how population migration and genetic admixture have shaped the current distribution and landscape of human kidney-associated diseases. Differences in exposures to infectious agents, environmental toxins, dietary components and climate also have the potential to influence the evolutionary genetics of kidneys. Of note, selective pressure on loci associated with kidney disease is often from non-kidney diseases, and thus it is important to understand how the link between genome-wide selected loci and kidney disease occurs in relation to secondary nephropathies.
Collapse
|
29
|
Costa L, Corre S, Michel V, Le Luel K, Fernandes J, Ziveri J, Jouvion G, Danckaert A, Mouchet N, Da Silva Barreira D, Torres J, Camorlinga M, D'Elios MM, Fiette L, De Reuse H, Galibert MD, Touati E. USF1 defect drives p53 degradation during Helicobacter pylori infection and accelerates gastric carcinogenesis. Gut 2020; 69:1582-1591. [PMID: 31822580 PMCID: PMC7456735 DOI: 10.1136/gutjnl-2019-318640] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 10/24/2019] [Accepted: 11/24/2019] [Indexed: 12/19/2022]
Abstract
OBJECTIVE Helicobacter pylori (Hp) is a major risk factor for gastric cancer (GC). Hp promotes DNA damage and proteasomal degradation of p53, the guardian of genome stability. Hp reduces the expression of the transcription factor USF1 shown to stabilise p53 in response to genotoxic stress. We investigated whether Hp-mediated USF1 deregulation impacts p53-response and consequently genetic instability. We also explored in vivo the role of USF1 in gastric carcinogenesis. DESIGN Human gastric epithelial cell lines were infected with Hp7.13, exposed or not to a DNA-damaging agent camptothecin (CPT), to mimic a genetic instability context. We quantified the expression of USF1, p53 and their target genes, we determined their subcellular localisation by immunofluorescence and examined USF1/p53 interaction. Usf1-/- and INS-GAS mice were used to strengthen the findings in vivo and patient data examined for clinical relevance. RESULTS In vivo we revealed the dominant role of USF1 in protecting gastric cells against Hp-induced carcinogenesis and its impact on p53 levels. In vitro, Hp delocalises USF1 into foci close to cell membranes. Hp prevents USF1/p53 nuclear built up and relocates these complexes in the cytoplasm, thereby impairing their transcriptional function. Hp also inhibits CPT-induced USF1/p53 nuclear complexes, exacerbating CPT-dependent DNA damaging effects. CONCLUSION Our data reveal that the depletion of USF1 and its de-localisation in the vicinity of cell membranes are essential events associated to the genotoxic activity of Hp infection, thus promoting gastric carcinogenesis. These findings are also of clinical relevance, supporting USF1 expression as a potential marker of GC susceptibility.
Collapse
Affiliation(s)
- Lionel Costa
- Unit of Helicobacter Pathogenesis, Department of Microbiology, CNRS ERL6002, Institut Pasteur, Paris, France,INSERM U1016, CNRS UMR 8104, Institut Cochin, Paris, France,Université Paris Diderot, Sorbone Paris Cité, Paris, France
| | - Sébastien Corre
- Institut de Génétique et Développement, Université de Rennes 1, Rennes, France
| | - Valérie Michel
- Unit of Helicobacter Pathogenesis, Department of Microbiology, CNRS ERL6002, Institut Pasteur, Paris, France
| | - Krysten Le Luel
- Unit of Helicobacter Pathogenesis, Department of Microbiology, CNRS ERL6002, Institut Pasteur, Paris, France,Université Paris Diderot, Sorbone Paris Cité, Paris, France
| | - Julien Fernandes
- Unit of Helicobacter Pathogenesis, Department of Microbiology, CNRS ERL6002, Institut Pasteur, Paris, France,UtechS PBI-C2RT, Institut Pasteur, Paris, France
| | - Jason Ziveri
- Unit of Helicobacter Pathogenesis, Department of Microbiology, CNRS ERL6002, Institut Pasteur, Paris, France,Pathogenesis of Systemic Infection, Institut Fédératif de Recherche Necker-Enfants Malades, Paris, France
| | - Gregory Jouvion
- Unit of Experimental Neuropathology, Department of Global Health, Institut Pasteur, Paris, France
| | | | - Nicolas Mouchet
- Institut de Génétique et Développement, Université de Rennes 1, Rennes, France
| | - David Da Silva Barreira
- Unit of Helicobacter Pathogenesis, Department of Microbiology, CNRS ERL6002, Institut Pasteur, Paris, France,AgroSup, Laboratoire PAM UMR A 02.102, Université de Bourgogne, Dijon, France
| | - Javier Torres
- Unidad de Investigación Médica en Enfermedades Infecciosas y Parasitarias, UMAE Pediatria, Instituto Mexicano del Seguro Social (IMSS), México city, Mexico
| | - Margarita Camorlinga
- Unidad de Investigación Médica en Enfermedades Infecciosas y Parasitarias, UMAE Pediatria, Instituto Mexicano del Seguro Social (IMSS), México city, Mexico
| | - Mario Milco D'Elios
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Laurence Fiette
- Unit of Experimental Neuropathology, Department of Global Health, Institut Pasteur, Paris, France,Institut Mutualiste Montsouris, Paris, France
| | - Hilde De Reuse
- Unit of Helicobacter Pathogenesis, Department of Microbiology, CNRS ERL6002, Institut Pasteur, Paris, France
| | - Marie-Dominique Galibert
- Institut de Génétique et Développement, Université de Rennes 1, Rennes, France,CHU, Department of Molecular Genetics and Genomics, Université de Rennes 1, Rennes, France
| | - Eliette Touati
- Unit of Helicobacter Pathogenesis, Department of Microbiology, CNRS ERL6002, Institut Pasteur, Paris, France
| |
Collapse
|
30
|
A novel protein upstream stimulatory factor 2 identified in lamprey, Lethenteron reissneri. Dev Genes Evol 2020; 230:347-357. [PMID: 32852621 DOI: 10.1007/s00427-020-00666-5] [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: 04/06/2020] [Accepted: 07/29/2020] [Indexed: 10/23/2022]
Abstract
Upstream stimulatory factors are kinds of multi-functional transcription factors, which are expressed in eukaryotes widely, including Upstream stimulatory factor 1 (USFl) and upstream stimulatory factor 2 (USF2). USF protein has a typical basic helix-loop-helix leucine zipper (b-HLH-LZ) structure, which is involved in cell cycle, cell proliferations, glucose and lipid metabolism, and other biochemical processes. Although the USF family is an important regulator of cellular processes, little is known about the USF genes of lampreys, especially their evolutionary relationships, expression profiles, and biological functions. Here, an upstream stimulatory factor 2 (USF2) homolog from lamprey (Lethenteron reissneri) was identified and characterized (designated as L-USF2) because it is closer to USF2 subfamily than to USF1 subfamily. The cDNA fragment of L-USF2 has an open reading frame (ORF) of 765-bp length, encodes 254 amino acids, and contains an HLH domain at the c-terminal of amino acids. Meanwhile, motifs and genetic structure analysis reveal that USF2 gene exons are conserved. Moreover, the 3D structure analysis indicates that L-USF2 adopts the general USF2 folding and has a high structural similarity with H-USF2. The synteny results showed that the L-USF2 adjacent gene changed greatly compared with the jaw vertebrates. By real-time quantitative experiment and Western blot analysis, we found that L-USF2 gene played a significant role in the immune responses. This study not only provides us with a further understanding of the evolution and function of the USF gene family but also provides a basis for exploring its immune responses and immune defenses in lampreys.
Collapse
|
31
|
Abstract
OBJECTIVES Chronic pancreatitis is the end stage of a pathologic inflammatory syndrome with multiple etiological factors, including genetic. We hypothesized that some pancreatitis etiology originates in pancreatic acinar or duct cells and requires both injury and compensatory mechanism failure. METHODS One hundred pancreatitis patients were assessed using a DNA sequencing panel for pancreatitis. Cooccurrence of variants within and between genes was measured. Gene coexpression was confirmed via published single-cell RNA sequencing. RESULTS One hundred and twenty-one variants were identified in 2 or more patients, 15 of which were enriched compared with reference populations. Single cell RNA-sequencing data verified coexpression of GGT1, CFTR, and PRSS1 in duct cells, PRSS1, CPA1, CEL, CTRC, and SPINK1 in acinar cells, and UBR1 in both. Multiple-risk variants with injury/stress effects (CEL, CFTR, CPA1, PRSS1) and impaired cell protection (CTRC, GGT1, SPINK1, UBR1) cooccur within duct cells, acinar cells, or both. CONCLUSIONS Pancreatitis is a complex disorder with genetic interactions across genes and cell types. These findings suggest a new, non-Mendelian genetic risk/etiology paradigm where a combination of nonpathogenic genetic risk variants in groups of susceptibility genes and injury/dysfunction response genes contribute to acquired pancreatic disease.
Collapse
|
32
|
Identifying Cattle Breed-Specific Partner Choice of Transcription Factors during the African Trypanosomiasis Disease Progression Using Bioinformatics Analysis. Vaccines (Basel) 2020; 8:vaccines8020246. [PMID: 32456126 PMCID: PMC7350023 DOI: 10.3390/vaccines8020246] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 05/13/2020] [Accepted: 05/21/2020] [Indexed: 12/18/2022] Open
Abstract
African Animal Trypanosomiasis (AAT) is a disease caused by pathogenic trypanosomes which affects millions of livestock every year causing huge economic losses in agricultural production especially in sub-Saharan Africa. The disease is spread by the tsetse fly which carries the parasite in its saliva. During the disease progression, the cattle are prominently subjected to anaemia, weight loss, intermittent fever, chills, neuronal degeneration, congestive heart failure, and finally death. According to their different genetic programs governing the level of tolerance to AAT, cattle breeds are classified as either resistant or susceptible. In this study, we focus on the cattle breeds N’Dama and Boran which are known to be resistant and susceptible to trypanosomiasis, respectively. Despite the rich literature on both breeds, the gene regulatory mechanisms of the underlying biological processes for their resistance and susceptibility have not been extensively studied. To address the limited knowledge about the tissue-specific transcription factor (TF) cooperations associated with trypanosomiasis, we investigated gene expression data from these cattle breeds computationally. Consequently, we identified significant cooperative TF pairs (especially DBP−PPARA and DBP−THAP1 in N’Dama and DBP−PAX8 in Boran liver tissue) which could help understand the underlying AAT tolerance/susceptibility mechanism in both cattle breeds.
Collapse
|
33
|
Yuan K, Wang D, Luan Q, Sun J, Gao Q, Jiang Z, Wang S, Han Y, Qu X, Cui Y, Qiu S, Di Y, Wang X, Song S, Wang P, Xia S, Yu Y, Liu W, Yin Y. Whole Genome Characterization and Genetic Evolution Analysis of a New Ostrich Parvovirus. Viruses 2020; 12:v12030334. [PMID: 32204363 PMCID: PMC7150892 DOI: 10.3390/v12030334] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 03/06/2020] [Accepted: 03/12/2020] [Indexed: 12/24/2022] Open
Abstract
Ostrich diseases characterized by paralysis have been breaking out in broad areas of China since 2015, causing major damage to the ostrich breeding industry in China. This report describes a parvovirus detected in ostriches from four different regions. The entire genomes of four parvovirus strains were sequenced following amplification by PCR, and we conducted comprehensive analysis of the ostrich parvovirus genome. Results showed that the length genomes of the parvovirus contained two open reading frames. Ostrich parvovirus (OsPV) is a branch of goose parvovirus (GPV). Genetic distance analysis revealed a close relationship between the parvovirus and goose parvovirus strains from China, with the closest being the 2016 goose parvovirus RC16 strain from Chongqing. This is the first report of a parvovirus in ostriches. However, whether OsPV is the pathogen of ostrich paralysis remains uncertain. This study contributes new information about the evolution and epidemiology of parvovirus in China, which provides a new way for the study of paralysis in ostriches.
Collapse
Affiliation(s)
- Kunpeng Yuan
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao 266019, China; (K.Y.); (Q.L.); (Q.G.); (Z.J.); (S.W.); (Y.H.); (X.Q.); (Y.C.); (S.Q.)
| | - Dongdong Wang
- Qingdao Bolong Experimental Animal Co., Ltd., Qingdao 266225, China; (D.W.); (J.S.)
| | - Qingdong Luan
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao 266019, China; (K.Y.); (Q.L.); (Q.G.); (Z.J.); (S.W.); (Y.H.); (X.Q.); (Y.C.); (S.Q.)
| | - Ju Sun
- Qingdao Bolong Experimental Animal Co., Ltd., Qingdao 266225, China; (D.W.); (J.S.)
| | - Qianwen Gao
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao 266019, China; (K.Y.); (Q.L.); (Q.G.); (Z.J.); (S.W.); (Y.H.); (X.Q.); (Y.C.); (S.Q.)
| | - Zhiyao Jiang
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao 266019, China; (K.Y.); (Q.L.); (Q.G.); (Z.J.); (S.W.); (Y.H.); (X.Q.); (Y.C.); (S.Q.)
| | - Shouchun Wang
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao 266019, China; (K.Y.); (Q.L.); (Q.G.); (Z.J.); (S.W.); (Y.H.); (X.Q.); (Y.C.); (S.Q.)
| | - Yijun Han
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao 266019, China; (K.Y.); (Q.L.); (Q.G.); (Z.J.); (S.W.); (Y.H.); (X.Q.); (Y.C.); (S.Q.)
| | - Xueting Qu
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao 266019, China; (K.Y.); (Q.L.); (Q.G.); (Z.J.); (S.W.); (Y.H.); (X.Q.); (Y.C.); (S.Q.)
| | - Yueying Cui
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao 266019, China; (K.Y.); (Q.L.); (Q.G.); (Z.J.); (S.W.); (Y.H.); (X.Q.); (Y.C.); (S.Q.)
| | - Shimei Qiu
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao 266019, China; (K.Y.); (Q.L.); (Q.G.); (Z.J.); (S.W.); (Y.H.); (X.Q.); (Y.C.); (S.Q.)
| | - Youxia Di
- China Ostrich Farming and Development Association, Beijing 100026, China; (Y.D.); (X.W.); (S.S.); (P.W.); (S.X.)
| | - Xiaoyi Wang
- China Ostrich Farming and Development Association, Beijing 100026, China; (Y.D.); (X.W.); (S.S.); (P.W.); (S.X.)
| | - Shige Song
- China Ostrich Farming and Development Association, Beijing 100026, China; (Y.D.); (X.W.); (S.S.); (P.W.); (S.X.)
| | - Peiheng Wang
- China Ostrich Farming and Development Association, Beijing 100026, China; (Y.D.); (X.W.); (S.S.); (P.W.); (S.X.)
| | - Shilong Xia
- China Ostrich Farming and Development Association, Beijing 100026, China; (Y.D.); (X.W.); (S.S.); (P.W.); (S.X.)
| | - Yongle Yu
- College of Biological Sciences, China Agricultural University, Beijing 100193, China;
| | - Weiquan Liu
- College of Biological Sciences, China Agricultural University, Beijing 100193, China;
- Correspondence: (W.L.); (Y.Y.)
| | - Yanbo Yin
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao 266019, China; (K.Y.); (Q.L.); (Q.G.); (Z.J.); (S.W.); (Y.H.); (X.Q.); (Y.C.); (S.Q.)
- Correspondence: (W.L.); (Y.Y.)
| |
Collapse
|
34
|
Irrelevance of USF2 rs916145 polymorphism with the risk of biliary atresia susceptibility in Southern Chinese children. Biosci Rep 2020; 40:222122. [PMID: 32109289 PMCID: PMC7048685 DOI: 10.1042/bsr20193623] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 02/06/2020] [Accepted: 02/13/2020] [Indexed: 02/07/2023] Open
Abstract
Backgrounds: Biliary atresia (BA) is a very rare neonatal disease, however, it has been the most common cause of obstructive jaundice in infancy. The complex pathogenesis of BA is not entirely clear and a lot of possible pathogenic mechanisms have been proposed to explain the etiology of BA, including genetic, inflammatory, environmental and developmental abnormalities. As a transcription factor, USF2 gene rs916145 polymorphism has been shown to be related to the risk of BA. Methods: We examined the USF2 rs916145 genotype in a large case–control study consisting of 506 BA patients and 1473 healthy controls, using the MassARRAY iPLEX Gold system (Sequenom). Odds ratios (ORs) and 95% confidence intervals (CIs) were used to evaluate the association between the USF2 gene rs916145 polymorphism and BA susceptibility. Results: The frequency of different genotypes showed no statistical significance (GG/GC, OR: 1.09, P=0.470, 95% CI: 0.87–1.35; GG/CC, OR: 0.86, P=0.378, 95% CI: 0.62–1.20). No obvious association was revealed between the USF2 gene rs916145 polymorphism and BA susceptibility. Conclusion:USF2 rs916145 polymorphism may not be the best predictor of BA.
Collapse
|
35
|
Nakayama T, Hirota T, Asaka D, Sakashita M, Ninomiya T, Morikawa T, Okano M, Haruna S, Yoshida N, Takeno S, Tanaka Y, Yoshikawa M, Ishitoya J, Hizawa N, Isogai S, Mitsui C, Taniguchi M, Kojima H, Fujieda S, Tamari M. A genetic variant near TSLP is associated with chronic rhinosinusitis with nasal polyps and aspirin-exacerbated respiratory disease in Japanese populations. Allergol Int 2020; 69:138-140. [PMID: 31326260 DOI: 10.1016/j.alit.2019.06.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Revised: 05/30/2019] [Accepted: 06/19/2019] [Indexed: 11/16/2022] Open
Affiliation(s)
- Tsuguhisa Nakayama
- Department of Otorhinolaryngology, The Jikei University School of Medicine, Tokyo, Japan; Laboratory for Respiratory and Allergic Diseases, Center for Integrative Medical Sciences, Institute of Physical and Chemical Research (RIKEN), Kanagawa, Japan
| | - Tomomitsu Hirota
- Laboratory for Respiratory and Allergic Diseases, Center for Integrative Medical Sciences, Institute of Physical and Chemical Research (RIKEN), Kanagawa, Japan; Division of Molecular Genetics, Research Center for Medical Science, The Jikei University School of Medicine, Tokyo, Japan
| | - Daiya Asaka
- Department of Otorhinolaryngology, The Jikei University School of Medicine, Tokyo, Japan
| | - Masafumi Sakashita
- Department of Otorhinolaryngology-Head and Neck Surgery, School of Medicine, University of Fukui, Fukui, Japan
| | - Takahiro Ninomiya
- Department of Otorhinolaryngology-Head and Neck Surgery, School of Medicine, University of Fukui, Fukui, Japan
| | - Taiyo Morikawa
- Department of Otorhinolaryngology-Head and Neck Surgery, School of Medicine, University of Fukui, Fukui, Japan
| | - Mitsuhiro Okano
- Department of Otolaryngology Head & Neck Surgery, Okayama University Graduate School of Medicine, Okayama, Japan
| | - Shinichi Haruna
- Department of Otorhinolaryngology Head & Neck Surgery, Dokkyo Medical University, Tochigi, Japan
| | - Naohiro Yoshida
- Department of Otolaryngology, Jichi Medical University Saitama Medical Center, Saitama, Japan
| | - Sachio Takeno
- Department of Otorhinolaryngology-Head & Neck Surgery, Graduate School of Biomedical Sciences, Hiroshima University, Hiroshima, Japan
| | - Yasuhiro Tanaka
- Department of Otolaryngology, Dokkyo Medical University, Koshigaya Hospital, Saitama, Japan
| | - Mamoru Yoshikawa
- Department of Otorhinolaryngology, Toho University Ohashi Medical Center, Tokyo, Japan
| | - Junichi Ishitoya
- Department of Otorhinolaryngology, Yokohama City University Medical Center, Kanagawa, Japan
| | - Nobuyuki Hizawa
- Division of Respiratory Medicine, Institute of Clinical Medicine, University of Tsukuba, Ibaraki, Japan
| | - Sumito Isogai
- Department of Respiratory Medicine, Fujita Health University, Aichi, Japan
| | - Chihiro Mitsui
- Clinical Research Center for Allergy and Rheumatology, Sagamihara National Hospital, Sagamihara, Kanagawa, Japan
| | - Masami Taniguchi
- Clinical Research Center for Allergy and Rheumatology, Sagamihara National Hospital, Sagamihara, Kanagawa, Japan
| | - Hiromi Kojima
- Department of Otorhinolaryngology, The Jikei University School of Medicine, Tokyo, Japan
| | - Shigeharu Fujieda
- Department of Otorhinolaryngology-Head and Neck Surgery, School of Medicine, University of Fukui, Fukui, Japan
| | - Mayumi Tamari
- Laboratory for Respiratory and Allergic Diseases, Center for Integrative Medical Sciences, Institute of Physical and Chemical Research (RIKEN), Kanagawa, Japan; Division of Molecular Genetics, Research Center for Medical Science, The Jikei University School of Medicine, Tokyo, Japan.
| |
Collapse
|
36
|
Ectopic Methylation of a Single Persistently Unmethylated CpG in the Promoter of the Vitellogenin Gene Abolishes Its Inducibility by Estrogen through Attenuation of Upstream Stimulating Factor Binding. Mol Cell Biol 2019; 39:MCB.00436-19. [PMID: 31548262 DOI: 10.1128/mcb.00436-19] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 09/15/2019] [Indexed: 01/02/2023] Open
Abstract
The enhancer/promoter of the vitellogenin II gene (VTG) has been extensively studied as a model system of vertebrate transcriptional control. While deletion mutagenesis and in vivo footprinting identified the transcription factor (TF) binding sites governing its tissue specificity, DNase hypersensitivity and DNA methylation studies revealed the epigenetic changes accompanying its hormone-dependent activation. Moreover, upon induction with estrogen (E2), the region flanking the estrogen-responsive element (ERE) was reported to undergo active DNA demethylation. We now show that although the VTG ERE is methylated in embryonic chicken liver and in LMH/2A hepatocytes, its induction by E2 was not accompanied by extensive demethylation. In contrast, E2 failed to activate a VTG enhancer/promoter-controlled luciferase reporter gene methylated by SssI. Surprisingly, this inducibility difference could be traced not to the ERE but rather to a single CpG in an E-box (CACGTG) sequence upstream of the VTG TATA box, which is unmethylated in vivo but methylated by SssI. We demonstrate that this E-box binds the upstream stimulating factor USF1/2. Selective methylation of the CpG within this binding site with an E-box-specific DNA methyltransferase, Eco72IM, was sufficient to attenuate USF1/2 binding in vitro and abolish the hormone-induced transcription of the VTG gene in the reporter system.
Collapse
|
37
|
Taghizadeh E, Mirzaei F, Jalilian N, Ghayour Mobarhan M, Ferns GA, Pasdar A. A novel mutation in
USF1
gene is associated with familial combined hyperlipidemia. IUBMB Life 2019; 72:616-623. [DOI: 10.1002/iub.2186] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Accepted: 10/01/2019] [Indexed: 12/30/2022]
Affiliation(s)
- Eskandar Taghizadeh
- Department of Medical Genetics, Faculty of MedicineMashhad University of Medical Sciences Mashhad Iran
- Cellular and Molecular Research CenterYasuj University of Medical Sciences Yasuj Iran
| | - Farzaneh Mirzaei
- Department of Medical Genetics, Faculty of MedicineMashhad University of Medical Sciences Mashhad Iran
| | - Nazanin Jalilian
- Department of Clinical biochemistry, School of MedicineKermanshah University of Medical Sciences Kermanshah Iran
| | - Majid Ghayour Mobarhan
- Metabolic Syndrome Research Centre, School of MedicineMashhad University of Medical Sciences Mashhad Iran
| | - Gordon A. Ferns
- Department of Medical EducationBrighton and Sussex Medical School Perso Falmer Brighton UK
| | - Alireza Pasdar
- Department of Medical Genetics, Faculty of MedicineMashhad University of Medical Sciences Mashhad Iran
- Medical Genetics Research Centre, Faculty of MedicineMashhad University of Medical Sciences Mashhad Iran
- Division of Applied Medicine, Medical School, University of Aberdeen Foresterhill Aberdeen UK
| |
Collapse
|
38
|
Tan X, Xu P, Zhang Y, Zhang PJ. Olive flounder (Paralichthys olivaceus) myogenic regulatory factor 4 and its muscle-specific promoter activity. Comp Biochem Physiol B Biochem Mol Biol 2019; 236:110310. [PMID: 31255700 DOI: 10.1016/j.cbpb.2019.110310] [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: 03/31/2019] [Revised: 06/21/2019] [Accepted: 06/24/2019] [Indexed: 02/07/2023]
Abstract
Myogenic regulatory factor 4 (MRF4) is a basic helix-loop-helix (bHLH) transcription factor that plays crucial roles in myoblast differentiation and maturation. Here, we report the isolation of the olive flounder (Paralichthys olivaceus) mrf4 gene and the spatiotemporal analysis of its expression patterns. Sequence analysis indicated that flounder mrf4 shared a similar structure with other vertebrate MRF4, including the conserved bHLH domain. Flounder mrf4 contains 3 exons and 2 introns. Sequence alignment and phylogenetic analysis showed that it was highly homologous with Salmo salar, Danio rerio, Takifugu rubripes, and Tetraodon nigroviridis mrf4. Flounder mrf4 was first expressed in the medial region of somites that give rise to slow muscles, and later spread to the lateral region of somites that give rise to fast muscles. Mrf4 transcript levels decreased significantly in mature somites in the trunk region, and expression could only be detected in the caudal somites, consistent with the timing of somite maturation. Transient expression analysis showed that the 506 bp flounder mrf4 promoter was sufficient to direct muscle-specific GFP expression in zebrafish embryos.
Collapse
Affiliation(s)
- Xungang Tan
- Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, PR China; Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266071, PR China.
| | - Peng Xu
- Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, PR China; Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266071, PR China; University of Chinese Academy of Sciences, Beijing 10049, PR China
| | - Yuqing Zhang
- Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, PR China; Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266071, PR China; University of Chinese Academy of Sciences, Beijing 10049, PR China
| | - Pei-Jun Zhang
- Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, PR China; Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266071, PR China
| |
Collapse
|
39
|
Liu F, Wang X, Zheng B, Li D, Chen C, Lee IS, Zhong J, Li D, Liu Y. USF2 enhances the osteogenic differentiation of PDLCs by promoting ATF4 transcriptional activities. J Periodontal Res 2019; 55:68-76. [PMID: 31448831 DOI: 10.1111/jre.12689] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Revised: 05/22/2019] [Accepted: 07/11/2019] [Indexed: 12/27/2022]
Abstract
OBJECTIVE Our study aimed to elucidate the regulatory molecules related to the osteogenic differentiation of periodontal ligament cells (PDLCs). BACKGROUND Periodontal ligament cells are a favorable source for cell-based therapy in periodontal bone engineering and regeneration due to their potential multilineage differentiation ability. However, the molecular mechanism and signaling pathways related to the osteogenic differentiation of PDLCs are still unclear. METHODS Osteoblast-specific protein expression levels were examined by ELISA in osteogenic-induced PDLCs (induced-PDLC group). A microarray assay and a bioinformatics analysis were carried out to reveal significantly expressed genes and the related pathways in induced-PDLCs, and these findings were then confirmed by qRT-PCR and a luciferase reporter assay. Finally, overexpressing and silencing gene systems were established to identify the specific transcriptional relationship and function of the target genes on the osteogenic differentiation of PDLCs. RESULTS Osteogenically differentiated PDLCs with high levels of osteoblast-specific proteins were established. The upstream stimulatory factor 2 (USF2) and activating transcription factor 4 (ATF4) mRNA levels were upregulated the most through the MAPK signaling pathway in the induced-PDLC group. USF2 could bind to the transcriptional initiation region of ATF4 and regulate its transcriptional activities. Additionally, the overexpression of USF2 promoted osteoblast-specific gene expression and the Alizarin red staining of PDLCs, while simultaneously overexpressing USF2 and silencing ATF4 reversed the favorable osteogenic effect of the induced-PDLCs by reducing osteoblast-specific gene expression and the Alizarin red staining level. CONCLUSION Our study demonstrated that USF2 could enhance the osteogenic differentiation of PDLCs by regulating ATF4 transcriptional activities, which provides a new strategy to utilize USF2 and ATF4 as potential target molecules for periodontal bone regeneration.
Collapse
Affiliation(s)
- Fan Liu
- Department of Orthodontics, School of Stomatology, China Medical University, Shenyang, China.,Department of Tissue Engineering, School of Fundamental Sciences, China Medical University, Shenyang, China
| | - Xiaohong Wang
- Department of Tissue Engineering, School of Fundamental Sciences, China Medical University, Shenyang, China
| | - Bowen Zheng
- Department of Orthodontics, School of Stomatology, China Medical University, Shenyang, China
| | - Danni Li
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, China
| | - Cen Chen
- College of Life Sciences, Zhejiang Sci-Tech University, Hangzhou, China
| | - In-Seop Lee
- Institute of Natural Science, Yonsei University, Seoul, Korea
| | - Jialin Zhong
- Key Laboratory of Oral Medicine, Guangzhou Institute of Oral Disease, Stomatology Hospital of Guangzhou Medical University, Guangzhou, China
| | - Duo Li
- Department of Orthodontics, School of Stomatology, China Medical University, Shenyang, China
| | - Yi Liu
- Department of Orthodontics, School of Stomatology, China Medical University, Shenyang, China
| |
Collapse
|
40
|
Xiaoli AM, Song Z, Yang F. Lipogenic SREBP-1a/c transcription factors activate expression of the iron regulator hepcidin, revealing cross-talk between lipid and iron metabolisms. J Biol Chem 2019; 294:12743-12753. [PMID: 31270208 DOI: 10.1074/jbc.ra119.009644] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 07/01/2019] [Indexed: 12/17/2022] Open
Abstract
The sterol regulatory element-binding proteins (SREBPs) are a family of transcription factors best known for stimulating the expression of genes encoding key lipogenic enzymes. However, SREBP functions beyond lipid metabolism are less understood. Here, we show that hepcidin antimicrobial peptide (Hamp), encoding the hormone hepcidin essential for iron homeostasis and regulated by dietary iron and inflammation, is a target gene of the two SREBP isoforms SREBP-1a/c. We found that in tissue culture, mature, active, and nuclear forms of the SREBP-1a/c proteins induce endogenous Hamp gene expression and increase the Hamp promoter activity primarily via three regulatory sequences, including an E-box. Moreover, ChIP experiments revealed that SREBP-1a binds to the Hamp gene promoter. Overexpression of nuclear SREBP-1a under the control of the phosphoenolpyruvate carboxylase-1 (Pck1) promoter in mice increased hepatic Hamp mRNA and blood hepcidin levels, and as expected, caused fatty liver. Consistent with the known effects of Hamp up-regulation, SREBP-1a-overexpressing mice displayed signs of dysregulation in iron metabolism, including reduced serum iron and increased hepatic and splenic iron storage. Conversely, liver-specific depletion of the nuclear forms of SREBPs, as in SREBP cleavage-activating protein knockout mice, impaired lipopolysaccharide-induced up-regulation of hepatic Hamp Together, these results indicate that the SREBP-1a/c transcription regulators activate hepcidin expression and thereby contribute to the control of mammalian iron metabolism.
Collapse
Affiliation(s)
- Alus M Xiaoli
- Departments of Medicine and Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, New York 10461
| | - Ziyi Song
- Departments of Medicine and Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, New York 10461
| | - Fajun Yang
- Departments of Medicine and Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, New York 10461
| |
Collapse
|
41
|
Tsang TF, Chan B, Tai WCS, Huang G, Wang J, Li X, Jiang ZH, Hsiao WLW. Gynostemma pentaphyllum saponins induce melanogenesis and activate cAMP/PKA and Wnt/β-catenin signaling pathways. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2019; 60:153008. [PMID: 31288940 DOI: 10.1016/j.phymed.2019.153008] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Accepted: 06/30/2019] [Indexed: 06/09/2023]
Abstract
BACKGROUND Melanogenesis is a physiological process of melanin production in response to UV exposure, which is modulated through multi-signaling pathways including cAMP/PKA, Wnt/β-catenin and MAPK signaling cascades. HYPOTHESIS/PURPOSE The present study aims to investigate the molecular mechanism of hyperpigmentation induced by Gynostemma pentaphyllum saponins. STUDY DESIGN/METHODS In this study, we investigated the melanogenic effects of triterpenoid saponins of Gynostemma pentaphyllum (GpS), a medicinal plant. Two mouse melanogenic cell lines B16 and B16F10 were employed for the current study. RESULTS The results showed that non-toxic doses of GpS markedly increased melanin formation in both B16 and B16F10 cells. Western blot analysis showed that GpS treatment significantly up-regulated the expression levels of the key melanogenic proteins, including tyrosinase (TYR), microphthalmia-associated transcription factor (MITF), TRP-1 and TRP-2 in a dose-dependent manner. The phospho-CREB, which is the downstream target of PKA is also elevated upon GpS treatment. We further observed that H89, a PKA inhibitor, attenuated the GpS induced tyrosinase activity, melanin content, the expression of phospho-CREB. In addition to the cAMP/PKA signaling pathway, GpS treatment also up-regulated the β-catenin of the Wnt signaling pathway which is involved in the transcriptional activation of MITF in melanogensis. We further demonstrated that treatment with GpS markedly enhance mRNA expression of MITF, along with the downstream target molecules, TYR, TRP-1 and TRP-2. Knock-down MITF with siMITF inhibited the expression of MITF mRNA by 63%, and the melanin content was reduced 70% in the siMITF-transfected cells compared to untransfected or scramble siRNA control cells. CONCLUSION These findings demonstrated strong melanogenic activities of GpS, and the MITF is essential for the melanogenesis stimulated by GpS.
Collapse
Affiliation(s)
- Ting-Fung Tsang
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Brandon Chan
- Department of Applied Biology Chemical Technology, The Hong Kong Polytechnic University, Hong Kong SAR, China
| | - William Chi-Shing Tai
- Department of Applied Biology Chemical Technology, The Hong Kong Polytechnic University, Hong Kong SAR, China; State Key Laboratory of Chinese Medicine and Molecular Pharmacology (Incubation), The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, 518057, China
| | - Guoxin Huang
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Jingrong Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Xiaoang Li
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Zhi Hong Jiang
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - W L Wendy Hsiao
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China.
| |
Collapse
|
42
|
Wang S, Xiao S, Cheng X, Chen S, Zhu X, Lin F, Chen S. Recovery of Muscovy duck-origin goose parvovirus from an infectious clone containing an E-box motif (CACATG) deletion within the left terminal region. Mol Cell Probes 2019; 46:101410. [PMID: 31128205 DOI: 10.1016/j.mcp.2019.05.006] [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: 11/27/2018] [Revised: 05/01/2019] [Accepted: 05/20/2019] [Indexed: 12/16/2022]
Abstract
Muscovy duck-origin goose parvovirus (MDGPV) is a causative agent of MDGPV-associated Derzsy's disease. To evalute the role of the cis-acting element E-box (CACATG) deletion on MDGPV eplication, an infectious plasmid clone p-PTΔE287, having one E-box deletion at nucleotide (nt) 287 of the left inverted terminal repeat sequence (L-ITR), was constructed by overlap extension PCR deleting the 287CACATG292 motif from the plasmid pMDGPVPT containing the full-length genome of the virulent MDGPV strain PT. The p-PTΔE287 plasmid was transfected into 9-day-old non-immune Muscovy duck embryos via the yolk sac, resulting in successful rescue of the deletion mutant virus r-PTΔE287. Compared with its parental virus PT, the virulence and the replication ability of r-PTΔE287 were reduced. In addition, we examined the ability of r-PTΔE287 to manipulate cell cycle progression. The results showed that r-PTΔE287 replication results in G0/G1 phase accumulation of infected duck embryo liver mesenchymal stem cells (BMSCs) and that this accumulation is caused by the prevention of cell cycle entry from G0/G1 phase into S phase. Taken together, introducing 287CACATG292 element deletion into MDGPV PT genomic DNA that induced rescued mutant virus (r-PTΔE287) cell cycle arrest function at the G0/G1 phase, which might inhibit MDGPV replication and virus progeny production. This study laid the foundation for further understanding of the relationship between E-box deletion in the L-ITR and MDGPV virulence.
Collapse
Affiliation(s)
- Shao Wang
- Institute of Animal Husbandry and Veterinary Medicine, Fujian Academy of Agricultural Sciences, Fuzhou, 350013, China; Fujian Animal Diseases Control Technology Development Center, Fuzhou, 350013, China
| | - Shifeng Xiao
- Institute of Animal Husbandry and Veterinary Medicine, Fujian Academy of Agricultural Sciences, Fuzhou, 350013, China; Fujian Animal Diseases Control Technology Development Center, Fuzhou, 350013, China
| | - Xiaoxia Cheng
- Institute of Animal Husbandry and Veterinary Medicine, Fujian Academy of Agricultural Sciences, Fuzhou, 350013, China; Fujian Animal Diseases Control Technology Development Center, Fuzhou, 350013, China
| | - Shaoying Chen
- Institute of Animal Husbandry and Veterinary Medicine, Fujian Academy of Agricultural Sciences, Fuzhou, 350013, China; Fujian Animal Diseases Control Technology Development Center, Fuzhou, 350013, China.
| | - Xiaoli Zhu
- Institute of Animal Husbandry and Veterinary Medicine, Fujian Academy of Agricultural Sciences, Fuzhou, 350013, China; Fujian Animal Diseases Control Technology Development Center, Fuzhou, 350013, China
| | - Fengqiang Lin
- Institute of Animal Husbandry and Veterinary Medicine, Fujian Academy of Agricultural Sciences, Fuzhou, 350013, China; Fujian Animal Diseases Control Technology Development Center, Fuzhou, 350013, China
| | - Shilong Chen
- Institute of Animal Husbandry and Veterinary Medicine, Fujian Academy of Agricultural Sciences, Fuzhou, 350013, China; Fujian Animal Diseases Control Technology Development Center, Fuzhou, 350013, China
| |
Collapse
|
43
|
Faisal I, Cisneros-Montalvo S, Hamer G, Tuominen MM, Laurila PP, Tumiati M, Jauhiainen M, Kotaja N, Toppari J, Mäkelä JA, Kauppi L. Transcription Factor USF1 Is Required for Maintenance of Germline Stem Cells in Male Mice. Endocrinology 2019; 160:1119-1136. [PMID: 30759202 DOI: 10.1210/en.2018-01088] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Accepted: 02/08/2019] [Indexed: 12/22/2022]
Abstract
A prerequisite for lifelong sperm production is that spermatogonial stem cells (SSCs) balance self-renewal and differentiation, yet factors required for this balance remain largely undefined. Using mouse genetics, we now demonstrate that the ubiquitously expressed transcription factor upstream stimulatory factor (USF)1 is critical for the maintenance of SSCs. We show that USF1 is not only detected in Sertoli cells as previously reported, but also in SSCs. Usf1-deficient mice display progressive spermatogenic decline as a result of age-dependent loss of SSCs. According to our data, the germ cell defect in Usf1-/- mice cannot be attributed to impairment of Sertoli cell development, maturation, or function, but instead is likely due to an inability of SSCs to maintain a quiescent state. SSCs of Usf1-/- mice undergo continuous proliferation, which provides an explanation for their age-dependent depletion. The proliferation-coupled exhaustion of SSCs in turn results in progressive degeneration of the seminiferous epithelium, gradual decrease in sperm production, and testicular atrophy. We conclude that the general transcription factor USF1 is indispensable for the proper maintenance of mammalian spermatogenesis.
Collapse
Affiliation(s)
- Imrul Faisal
- Genome-Scale Biology Research Program, Research Programs Unit, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Department of Biochemistry and Developmental Biology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Doctoral Program in Biomedicine, Doctoral School in Health Sciences, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Sheyla Cisneros-Montalvo
- Institute of Biomedicine, Research Centre for Integrative Physiology and Pharmacology, University of Turku, Turku, Finland
- Turku Doctoral Program of Molecular Medicine, University of Turku, Turku, Finland
| | - Geert Hamer
- Center for Reproductive Medicine, Amsterdam Research Institute Reproduction and Development, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Minna M Tuominen
- Genome-Scale Biology Research Program, Research Programs Unit, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Pirkka-Pekka Laurila
- Genomics and Biomarkers Unit, National Institute for Health and Welfare, Biomedicum, Helsinki, Finland
- Department of Medical and Clinical Genetics, University of Helsinki, Helsinki, Finland
| | - Manuela Tumiati
- Genome-Scale Biology Research Program, Research Programs Unit, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Matti Jauhiainen
- Genomics and Biomarkers Unit, National Institute for Health and Welfare, Biomedicum, Helsinki, Finland
- Minerva Foundation Institute for Medical Research, Helsinki, Finland
| | - Noora Kotaja
- Institute of Biomedicine, Research Centre for Integrative Physiology and Pharmacology, University of Turku, Turku, Finland
| | - Jorma Toppari
- Institute of Biomedicine, Research Centre for Integrative Physiology and Pharmacology, University of Turku, Turku, Finland
- Department of Pediatrics, Turku University Hospital, Turku, Finland
| | - Juho-Antti Mäkelä
- Institute of Biomedicine, Research Centre for Integrative Physiology and Pharmacology, University of Turku, Turku, Finland
| | - Liisa Kauppi
- Genome-Scale Biology Research Program, Research Programs Unit, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Department of Biochemistry and Developmental Biology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| |
Collapse
|
44
|
Chi TF, Horbach T, Götz C, Kietzmann T, Dimova EY. Cyclin-Dependent Kinase 5 (CDK5)-Mediated Phosphorylation of Upstream Stimulatory Factor 2 (USF2) Contributes to Carcinogenesis. Cancers (Basel) 2019; 11:cancers11040523. [PMID: 31013770 PMCID: PMC6521020 DOI: 10.3390/cancers11040523] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2019] [Revised: 03/30/2019] [Accepted: 04/08/2019] [Indexed: 12/12/2022] Open
Abstract
The transcription factor USF2 is supposed to have an important role in tumor development. However, the regulatory mechanisms contributing to the function of USF2 are largely unknown. Cyclin-dependent kinase 5 (CDK5) seems to be of importance since high levels of CDK5 were found in different cancers associated with high USF2 expression. Here, we identified USF2 as a phosphorylation target of CDK5. USF2 is phosphorylated by CDK5 at two serine residues, serine 155 and serine 222. Further, phosphorylation of USF2 at these residues was shown to stabilize the protein and to regulate cellular growth and migration. Altogether, these results delineate the importance of the CDK5-USF2 interplay in cancer cells.
Collapse
Affiliation(s)
- Tabughang Franklin Chi
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, 90014 Oulu, Finland; (T.F.C.); (T.K.)
| | - Tina Horbach
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, 90014 Oulu, Finland; (T.F.C.); (T.K.)
| | - Claudia Götz
- Medical Biochemistry and Molecular Biology, Saarland University, 66421 Homburg, Germany;
| | - Thomas Kietzmann
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, 90014 Oulu, Finland; (T.F.C.); (T.K.)
| | - Elitsa Y. Dimova
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, 90014 Oulu, Finland; (T.F.C.); (T.K.)
- Correspondence: ; Tel.: +358-0-294-485-785; Fax: +358-8-553-114
| |
Collapse
|
45
|
Muhammad JS, Eladl MA, Khoder G. Helicobacter pylori-induced DNA Methylation as an Epigenetic Modulator of Gastric Cancer: Recent Outcomes and Future Direction. Pathogens 2019; 8:23. [PMID: 30781778 PMCID: PMC6471032 DOI: 10.3390/pathogens8010023] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Revised: 02/04/2019] [Accepted: 02/11/2019] [Indexed: 02/07/2023] Open
Abstract
Gastric cancer is ranked fifth in cancer list and has the third highest mortality rate. Helicobacter pylori is a class I carcinogen and a predominant etiological factor of gastric cancer. H. pylori infection may induce carcinogenesis via epigenetic alterations in the promoter region of various genes. H. pylori is known to induce hypermethylation-silencing of several tumor suppressor genes in H. pylori-infected cancerous and H. pylori-infected non-cancerous gastric mucosae. This article presents a review of the published literature mainly from the last year 15 years. The topic focuses on H. pylori-induced DNA methylation linked to gastric cancer development. The authors have used MeSH terms "Helicobacter pylori" with "epigenetic," "DNA methylation," in combination with "gastric inflammation", gastritis" and "gastric cancer" to search SCOPUS, PubMed, Ovid, and Web of Science databases. The success of epigenetic drugs such as de-methylating agents in the treatment of certain cancers has led towards new prospects that similar approaches could also be applied against gastric cancer. However, it is very important to understand the role of all the genes that have already been linked to H. pylori-induced DNA methylation in order to in order to evaluate the potential benefits of epigenetic drugs.
Collapse
Affiliation(s)
- Jibran Sualeh Muhammad
- Department of Basic Medical Sciences, College of Medicine, University of Sharjah, Sharjah 27272, UAE.
| | - Mohamed Ahmed Eladl
- Department of Basic Medical Sciences, College of Medicine, University of Sharjah, Sharjah 27272, UAE.
| | - Ghalia Khoder
- Department of Pharmaceutics and Pharmaceutical Technology, College of Pharmacy, University of Sharjah, Sharjah 27272, UAE.
| |
Collapse
|
46
|
Cai ZN, Li W, Mehmood S, Pan WJ, Wu QX, Chen Y, Lu YM. Effect of polysaccharide FMP-1 from Morchella esculenta on melanogenesis in B16F10 cells and zebrafish. Food Funct 2019; 9:5007-5015. [PMID: 30188555 DOI: 10.1039/c8fo01267a] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Polysaccharides from Morchella esculenta are known to exhibit diverse bioactivities, while an anti-melanogenesis effect has been barely addressed. Herein, the anti-melanogenesis activity of a heteropolysaccharide from M. esculenta (FMP-1) was investigated in vitro and in vivo. FMP-1 had no significant cytotoxic effect on B16F10 melanoma cells as well as zebrafish larvae, but did reduce melanin contents and tyrosinase activities in both of them. Treatment with FMP-1 also effectively suppressed the expression of melanogenesis-related proteins, including MC1R, MITF, TRP-1 and TRP-2, through decreasing the phosphorylation of cyclic adenosine monophosphate response element-binding protein (CREB). Moreover, the mitogen-activated protein kinase (MAPK) pathway was observed mediating FMP-1's inhibitory effect against melanin production. Specifically, FMP-1 treatment markedly inhibited the activation of phosphorylation of p38 mitogen-activated protein kinase. These results suggested that FMP-1's inhibitory effect against melanogenesis is mediated by the inhibition of CREB and p38 signaling pathways, thereby resulting in the downstream repression of melanogenesis-related proteins and the subsequent melanin production. These data provide insight into FMP-1's potential anti-melanogenesis effect in food and cosmetic industries.
Collapse
Affiliation(s)
- Zheng-Nan Cai
- School of Life Sciences and Anhui Key Laboratory of Modern Biomanufacturing, Anhui University, Hefei 230601, Anhui, P. R. China.
| | | | | | | | | | | | | |
Collapse
|
47
|
Lv S, Fan H, Li J, Yang H, Huang J, Shu X, Zhang L, Xu Y, Li X, Zuo J, Xiao C. Genetic Polymorphisms of TYMS, MTHFR, ATIC, MTR, and MTRR Are Related to the Outcome of Methotrexate Therapy for Rheumatoid Arthritis in a Chinese Population. Front Pharmacol 2018; 9:1390. [PMID: 30546311 PMCID: PMC6279856 DOI: 10.3389/fphar.2018.01390] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2018] [Accepted: 11/12/2018] [Indexed: 11/22/2022] Open
Abstract
Objective: Analysis of the relationship between single nucleotide polymorphisms (SNPs) and outcomes of methotrexate (MTX) therapy for rheumatoid arthritis (RA) in China. Materials and Methods: TYMS 28 bp VNTR (rs34743033), MTHFR [677C>T (rs1801133) and 1298A>C (rs1801131)], ATIC 347C>G (rs2372536), MTR A2756G (rs1805087), and MTRR 66A>G (rs1801394) enzyme proteins may be related to the outcomes of MTX therapy, according to our previous meta-analysis. A total of 162 patients with RA were included in our study. SNPs were evaluated using polymerase chain reaction (PCR). Disease Activity Score 28 (DAS28) was used to evaluate the clinical response, and adverse drug reactions (ADRs) were collected after physical examinations of the patients. Results: The MTHFR 677C>T gene showed a relationship with the ADRs of MTX in the Recessive model [TT vs. (CC+CT)] (p = 0.04, OR = 2.20, 95% CI: 1.01, 4.77). In the Codominant model [CT vs. (CC+TT)], the MTHFR 677C>T gene also showed a trend of association with ADRs (p = 0.08, OR = 0.52, 95% CI: 0.25, 1.08). No significant difference was found between TYMS, MTHFR, ATIC, MTR, and MTRR gene polymorphisms and the RA response or ADRs related to MTX in our study. Conclusion: Our results showed that the MTHFR [677C>T (rs1801133)] TT genotype is associated with ADRs to MTX in Chinese RA patients. Other SNPs, including TYMS 28bp VNTR (rs34743033), MTHFR [677C>T (rs1801133) and 1298A>C (rs1801131)], ATIC 347C>G (rs2372536), MTR A2756G (rs1805087), and MTRR 66A>G (rs1801394) gene polymorphisms, were not associated with MTX treatment outcomes. Further studies are required to validate these findings.
Collapse
Affiliation(s)
- Shuang Lv
- Institute of Clinical Medicine, China-Japan Friendship Hospital, Beijing, China.,Beijing University of Chinese Medicine, Beijing, China
| | - HuiZhen Fan
- Department of Gastroenterology, People's Hospital of Yichun, Yichun, China
| | - Jiang Li
- Department of Laboratory Medicine, China-Japan Friendship Hospital, Beijing, China
| | - Hui Yang
- Department of Laboratory Medicine, China-Japan Friendship Hospital, Beijing, China
| | - Jing Huang
- Institute of Clinical Medicine, China-Japan Friendship Hospital, Beijing, China.,Beijing University of Chinese Medicine, Beijing, China
| | - XiaoMing Shu
- Department of Rheumatology, China-Japan Friendship Hospital, Beijing, China
| | - Lu Zhang
- Department of Rheumatology, China-Japan Friendship Hospital, Beijing, China
| | - Yuan Xu
- Department of TCM Rheumatology, China-Japan Friendship Hospital, Beijing, China
| | - Xiaoya Li
- Institute of Clinical Medicine, China-Japan Friendship Hospital, Beijing, China.,Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jieyu Zuo
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada
| | - Cheng Xiao
- Institute of Clinical Medicine, China-Japan Friendship Hospital, Beijing, China.,Beijing University of Chinese Medicine, Beijing, China.,Department of Gastroenterology, People's Hospital of Yichun, Yichun, China
| |
Collapse
|
48
|
OCT3 promoter haplotype is associated with metformin pharmacokinetics in Koreans. Sci Rep 2018; 8:16965. [PMID: 30446679 PMCID: PMC6240047 DOI: 10.1038/s41598-018-35322-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Accepted: 10/31/2018] [Indexed: 12/13/2022] Open
Abstract
Organic cation transporter 3 (OCT3) is expressed in various organs in humans and plays an important role in the transport of organic cations and drugs including metformin. In this study, we identified genetic variations of the OCT3 promoter and functionally characterized each variant by in vitro assays. Next, the association between the functional haplotype of the OCT3 promoter and pharmacokinetics of metformin was evaluated. In our study population, 7 variations and 2 major haplotypes were identified, of which H2 haplotype yielded a significantly higher luciferase activity than did the wild type. Two variants of H2, c.-1603G > A and c.-1547T > G, yielded significantly lower luciferase activities, whereas the luciferase activity of another variant, c.-29G > A, was significantly higher. Two transcription factors, Sp1 and USF1, were involved in the regulation of OCT3 transcription. Analysis of clinical data revealed that 25 subjects, either homozygous or heterozygous for H2, showed increased AUCinf and Cmax by 17.2% and 15.9%, respectively [P = 0.016 and 0.031, GMR (90% CI) = 1.17 (1.06–1.29) and 1.17 (1.04–1.31), respectively], compared to the 20 subjects in the control group. Our study suggests that an OCT3 promoter haplotype affects the pharmacokinetics of metformin in Koreans as well as the OCT3 transcription rate.
Collapse
|
49
|
Tan Y, Chen Y, Du M, Peng Z, Xie P. USF2 inhibits the transcriptional activity of Smurf1 and Smurf2 to promote breast cancer tumorigenesis. Cell Signal 2018; 53:49-58. [PMID: 30244169 DOI: 10.1016/j.cellsig.2018.09.013] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2018] [Revised: 09/16/2018] [Accepted: 09/19/2018] [Indexed: 10/28/2022]
Abstract
Smurf1 (Smad ubiquitylation regulatory factor 1) and Smurf2 are negative regulators of the TGF-β (transforming growth factor-β) pathway. The protein stability and ubiquitin E3 activity regulation of Smurfs have been well studied. However, the mechanism of Smurfs expression at the transcriptional level remains uncharacterized. Here, we reported that USF2 (upstream stimulatory factor 2), a basic helix-loop-helix-leucine-zip transcription factor, is necessary for the transcriptional activity of Smurf1 and Smurf2. The 5'-flanking sequences of the Smurfs gene have more than one E-box motifs, and USF2 bounds the Smurfs promoter in vitro and in vivo. Over-expression USF2 inhibited the transcriptional activity of the Smurfs, and Smurfs mRNA was markedly decreased. Therefore, the activity of TGF-β was distinctly enhanced. Furthermore, in human breast cancers, USF2 was abnormally high expressed and correlated with cancer progression. USF2 was specifically inversely correlated with Smurfs in Luminal A subtype breast cancer patients. These findings suggest the mechanism regulation of Smurfs transcriptional activity, and shed new light on the cancer-promoting role of USF2.
Collapse
Affiliation(s)
- Yawen Tan
- Department of Breast and Thyroid Surgery, The Second People's Hospital of Shenzhen, Guangdong 518035, China
| | - Yujiao Chen
- Department of Biochemistry and Molecular Biology, Capital Medical University, Beijing 100069, China; Beijing Key Laboratory for Cancer Invasion and Metastasis Research, Capital Medical University, Beijing 100069, China
| | - Mengge Du
- Department of Biochemistry and Molecular Biology, Capital Medical University, Beijing 100069, China; Beijing Key Laboratory for Cancer Invasion and Metastasis Research, Capital Medical University, Beijing 100069, China
| | - Zhiqiang Peng
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center of Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China.
| | - Ping Xie
- Department of Biochemistry and Molecular Biology, Capital Medical University, Beijing 100069, China; Beijing Key Laboratory for Cancer Invasion and Metastasis Research, Capital Medical University, Beijing 100069, China.
| |
Collapse
|
50
|
Wang S, Xiao S, Cheng X, Chen S, Zhu X, Lin F, Chen S. Construction and rescue of Muscovy duck-origin goose parvovirus from an infectious clone containing an E-box deletion within the left terminal region. Mol Cell Probes 2018; 42:32-35. [PMID: 30240819 DOI: 10.1016/j.mcp.2018.09.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2018] [Revised: 09/07/2018] [Accepted: 09/17/2018] [Indexed: 01/24/2023]
Abstract
To obtain a deletion mutant of Muscovy duck-origin goose parvovirus (MDGPV) and to analyze its biological characteristics, the pMDGPVPT plasmid, which contains a full-length DNA infectious clone of the MDGPV PT strain, was used in this study as the template. The E-box at nt 315 of the left inverted terminal repeat sequence (L-ITR) was deleted by overlap extension PCR to obtain the infectious recombinant plasmid p-PTΔE315. The p-PTΔE315 plasmid was transfected into 9-day-old non-immune Muscovy duck embryos via the yolk sac and the rescued deletion mutant virus r-PTΔE315 was generated. Experiments to demonstrate the novel deletion mutant virus' biological characteristics showed that r-PTΔE315 can cause typical lesions after infection of Muscovy duck embryos. Compared with its parent strain PT, the virulence of r-PTΔE315 and its proliferation ability in Muscovy duck embryos were attenuated, but its ability to replicate in MDEF cells was enhanced. This study laid the foundation for further understanding of the relationship between E-box deletion in the L-ITR and MDGPV virulence.
Collapse
Affiliation(s)
- Shao Wang
- Institute of Animal Husbandry and Veterinary Medicine, Fujian Academy of Agricultural Sciences, Fuzhou, 350013, China; Fujian Animal Diseases Control Technology Development Center, Fuzhou, 350013, China
| | - Shifeng Xiao
- Institute of Animal Husbandry and Veterinary Medicine, Fujian Academy of Agricultural Sciences, Fuzhou, 350013, China; Fujian Animal Diseases Control Technology Development Center, Fuzhou, 350013, China
| | - Xiaoxia Cheng
- Institute of Animal Husbandry and Veterinary Medicine, Fujian Academy of Agricultural Sciences, Fuzhou, 350013, China; Fujian Animal Diseases Control Technology Development Center, Fuzhou, 350013, China
| | - Shaoying Chen
- Institute of Animal Husbandry and Veterinary Medicine, Fujian Academy of Agricultural Sciences, Fuzhou, 350013, China; Fujian Animal Diseases Control Technology Development Center, Fuzhou, 350013, China.
| | - Xiaoli Zhu
- Institute of Animal Husbandry and Veterinary Medicine, Fujian Academy of Agricultural Sciences, Fuzhou, 350013, China; Fujian Animal Diseases Control Technology Development Center, Fuzhou, 350013, China
| | - Fengqiang Lin
- Institute of Animal Husbandry and Veterinary Medicine, Fujian Academy of Agricultural Sciences, Fuzhou, 350013, China; Fujian Animal Diseases Control Technology Development Center, Fuzhou, 350013, China
| | - Shilong Chen
- Institute of Animal Husbandry and Veterinary Medicine, Fujian Academy of Agricultural Sciences, Fuzhou, 350013, China; Fujian Animal Diseases Control Technology Development Center, Fuzhou, 350013, China
| |
Collapse
|