751
|
Lee SS, Lee SJ, Lee SH, Ryu JM, Lim HS, Kim JS, Song EJ, Jung YH, Lee HJ, Kim CH, Han HJ. Netrin-1-Induced Stem Cell Bioactivity Contributes to the Regeneration of Injured Tissues via the Lipid Raft-Dependent Integrin α6β4 Signaling Pathway. Sci Rep 2016; 6:37526. [PMID: 27881869 PMCID: PMC5121594 DOI: 10.1038/srep37526] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Accepted: 10/31/2016] [Indexed: 11/17/2022] Open
Abstract
Netrin-1 (Ntn-1) is a multifunctional neuronal signaling molecule; however, its physiological significance, which improves the tissue-regeneration capacity of stem cells, has not been characterized. In the present study, we investigate the mechanism by which Ntn-1 promotes the proliferation of hUCB-MSCs with regard to the regeneration of injured tissues. We found that Ntn-1 induces the proliferation of hUCB-MSCs mainly via Inα6β4 coupled with c-Src. Ntn-1 induced the recruitment of NADPH oxidases and Rac1 into membrane lipid rafts to facilitate ROS production. The Inα6β4 signaling of Ntn-1 through ROS production is uniquely mediated by the activation of SP1 for cell cycle progression and the transcriptional occupancy of SP1 on the VEGF promoter. Moreover, Ntn-1 has the ability to induce the F-actin reorganization of hUCB-MSCs via the Inα6β4 signaling pathway. In an in vivo model, transplantation of hUCB-MSCs pre-treated with Ntn-1 enhanced the skin wound healing process, where relatively more angiogenesis was detected. The potential effect of Ntn-1 on angiogenesis is further verified by the mouse hindlimb ischemia model, where the pre-activation of hUCB-MSCs with Ntn-1 significantly improved vascular regeneration. These results demonstrate that Ntn-1 plays an important role in the tissue regeneration process of hUCB-MSC via the lipid raft-mediated Inα6β4 signaling pathway.
Collapse
Affiliation(s)
- Soo Sang Lee
- Department of plastic and reconstructive surgery, Bundang CHA Medical Center, Yatap-dong, Bundang-gu, Seongnam-si, Gyeonggi-do, Korea.,SKY plastic surgery clinic, 4F, 826-23, Yeoksam-dong, Gangnam-gu, Seoul, Korea
| | - Sei-Jung Lee
- Department of Veterinary Physiology, College of Veterinary Medicine, Research Institute for Veterinary Science and BK21 PLUS Program for Creative Veterinary Science Research Center, Seoul National University, Seoul 08826, Korea
| | - Sang Hun Lee
- Medical Science Research Institute, Soonchunhyang University Seoul Hospital, Seoul, Korea
| | - Jung Min Ryu
- Department of Veterinary Physiology, College of Veterinary Medicine, Chonnam National University, Gwangju, Republic of Korea
| | - Hyeon Su Lim
- Department of Veterinary Physiology, College of Veterinary Medicine, Research Institute for Veterinary Science and BK21 PLUS Program for Creative Veterinary Science Research Center, Seoul National University, Seoul 08826, Korea
| | - Jun Sung Kim
- Department of Veterinary Physiology, College of Veterinary Medicine, Research Institute for Veterinary Science and BK21 PLUS Program for Creative Veterinary Science Research Center, Seoul National University, Seoul 08826, Korea
| | - Eun Ju Song
- Department of Veterinary Physiology, College of Veterinary Medicine, Research Institute for Veterinary Science and BK21 PLUS Program for Creative Veterinary Science Research Center, Seoul National University, Seoul 08826, Korea
| | - Young Hyun Jung
- Department of Veterinary Physiology, College of Veterinary Medicine, Research Institute for Veterinary Science and BK21 PLUS Program for Creative Veterinary Science Research Center, Seoul National University, Seoul 08826, Korea
| | - Hyun Jik Lee
- Department of Veterinary Physiology, College of Veterinary Medicine, Research Institute for Veterinary Science and BK21 PLUS Program for Creative Veterinary Science Research Center, Seoul National University, Seoul 08826, Korea
| | - Chung Hun Kim
- Department of plastic and reconstructive surgery, Bundang CHA Medical Center, Yatap-dong, Bundang-gu, Seongnam-si, Gyeonggi-do, Korea
| | - Ho Jae Han
- Department of Veterinary Physiology, College of Veterinary Medicine, Research Institute for Veterinary Science and BK21 PLUS Program for Creative Veterinary Science Research Center, Seoul National University, Seoul 08826, Korea
| |
Collapse
|
752
|
Pu J, Sun H, Wang J, Wu M, Wang K, Denholm I, Han Z. Multiple cis-acting elements involved in up-regulation of a cytochrome P450 gene conferring resistance to deltamethrin in smal brown planthopper, Laodelphax striatellus (Fallén). INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2016; 78:20-28. [PMID: 27590347 DOI: 10.1016/j.ibmb.2016.08.008] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Revised: 08/02/2016] [Accepted: 08/26/2016] [Indexed: 06/06/2023]
Abstract
As well as arising from single point mutations in binding sites or detoxifying enzymes, it is likely that insecticide resistance mechanisms are frequently controlled by multiple genetic factors, resulting in resistance being inherited as a quantitative trait. However, empirical evidence for this is still rare. Here we analyse the causes of up-regulation of CYP6FU1, a monoxygenase implicated in resistance to deltamethrin in the rice pest Laodelphax striatellus. The 5'-flanking region of this gene was cloned and sequenced from individuals of a susceptible and a resistant strain. A luminescent reporter assay was used to evaluate different 5'-flanking regions and their fragments for promoter activity. Mutations enhancing promoter activity in various fragments were characterized, singly and in combination, by site mutation recovery. Nucleotide diversity in flanking sequences was greatly reduced in deltamethrin-resistant insects compared to susceptible ones. Phylogenetic sequence analysis found that CYP6FU1 had five different types of 5'-flanking region. All five types were present in a susceptible strain but only a single type showing the highest promoter activity was present in a resistant strain. Four cis-acting elements were identified whose influence on up-regulation was much more pronounced in combination than when present singly. Of these, two were new transcription factor (TF) binding sites produced by mutations, another one was also a new TF binding site alternated from an existing one, and the fourth was a unique transcription start site. These results demonstrate that multiple cis-acting elements are involved in up-regulating CYP6FU1 to generate a resistance phenotype.
Collapse
Affiliation(s)
- Jian Pu
- Department of Entomology, College of Plant Protection, Nanjing Agricultural University, Jiangsu, The Key Laboratory of Monitoring and Management of Plant Diseases and Insects, Ministry of Agriculture, Nanjing, 210095, Jiangsu, China
| | - Haina Sun
- Department of Entomology, College of Plant Protection, Nanjing Agricultural University, Jiangsu, The Key Laboratory of Monitoring and Management of Plant Diseases and Insects, Ministry of Agriculture, Nanjing, 210095, Jiangsu, China
| | - Jinda Wang
- Department of Entomology, College of Plant Protection, Nanjing Agricultural University, Jiangsu, The Key Laboratory of Monitoring and Management of Plant Diseases and Insects, Ministry of Agriculture, Nanjing, 210095, Jiangsu, China
| | - Min Wu
- Department of Entomology, College of Plant Protection, Nanjing Agricultural University, Jiangsu, The Key Laboratory of Monitoring and Management of Plant Diseases and Insects, Ministry of Agriculture, Nanjing, 210095, Jiangsu, China
| | - Kangxu Wang
- Department of Entomology, College of Plant Protection, Nanjing Agricultural University, Jiangsu, The Key Laboratory of Monitoring and Management of Plant Diseases and Insects, Ministry of Agriculture, Nanjing, 210095, Jiangsu, China
| | - Ian Denholm
- Biological and Environmental Sciences Department, University of Hertfordshire, Hatfield, Hertfordshire, United Kingdom
| | - Zhaojun Han
- Department of Entomology, College of Plant Protection, Nanjing Agricultural University, Jiangsu, The Key Laboratory of Monitoring and Management of Plant Diseases and Insects, Ministry of Agriculture, Nanjing, 210095, Jiangsu, China.
| |
Collapse
|
753
|
Agrawal R, Dale TP, Al-Zubaidi MA, Benny Malgulwar P, Forsyth NR, Kulshreshtha R. Pluripotent and Multipotent Stem Cells Display Distinct Hypoxic miRNA Expression Profiles. PLoS One 2016; 11:e0164976. [PMID: 27783707 PMCID: PMC5081191 DOI: 10.1371/journal.pone.0164976] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Accepted: 10/04/2016] [Indexed: 12/20/2022] Open
Abstract
MicroRNAs are reported to have a crucial role in the regulation of self-renewal and differentiation of stem cells. Hypoxia has been identified as a key biophysical element of the stem cell culture milieu however, the link between hypoxia and miRNA expression in stem cells remains poorly understood. We therefore explored miRNA expression in hypoxic human embryonic and mesenchymal stem cells (hESCs and hMSCs). A total of 50 and 76 miRNAs were differentially regulated by hypoxia (2% O2) in hESCs and hMSCs, respectively, with a negligible overlap of only three miRNAs. We found coordinate regulation of precursor and mature miRNAs under hypoxia suggesting their regulation mainly at transcriptional level. Hypoxia response elements were located upstream of 97% of upregulated hypoxia regulated miRNAs (HRMs) suggesting hypoxia-inducible-factor (HIF) driven transcription. HIF binding to the candidate cis-elements of specific miRNAs under hypoxia was confirmed by Chromatin immunoprecipitation coupled with qPCR. Role analysis of a subset of upregulated HRMs identified linkage to reported inhibition of differentiation while a downregulated subset of HRMs had a putative role in the promotion of differentiation. MiRNA-target prediction correlation with published hypoxic hESC and hMSC gene expression profiles revealed HRM target genes enriched in the cytokine:cytokine receptor, HIF signalling and pathways in cancer. Overall, our study reveals, novel and distinct hypoxia-driven miRNA signatures in hESCs and hMSCs with the potential for application in optimised culture and differentiation models for both therapeutic application and improved understanding of stem cell biology.
Collapse
Affiliation(s)
- Rahul Agrawal
- Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology, Delhi, India-110016
| | - Tina P. Dale
- Guy Hilton Research Centre, Institute of Science and Technology in Medicine, University of Keele, Thornburrow Drive, Hartshill, Stoke-on-Trent, Staffordshire, ST4 7QB, United Kingdom
| | - Mohammed A. Al-Zubaidi
- Guy Hilton Research Centre, Institute of Science and Technology in Medicine, University of Keele, Thornburrow Drive, Hartshill, Stoke-on-Trent, Staffordshire, ST4 7QB, United Kingdom
- College of Pharmacy, Al-Mustansiriyah University, Baghdad, Iraq
| | - Prit Benny Malgulwar
- Department of Pathology, All India Institute of Medical Sciences, New Delhi, India-110029
| | - Nicholas R. Forsyth
- Guy Hilton Research Centre, Institute of Science and Technology in Medicine, University of Keele, Thornburrow Drive, Hartshill, Stoke-on-Trent, Staffordshire, ST4 7QB, United Kingdom
| | - Ritu Kulshreshtha
- Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology, Delhi, India-110016
| |
Collapse
|
754
|
Wang Y, Fang R, Cui M, Zhang W, Bai X, Wang H, Liu B, Zhang X, Ye L. The oncoprotein HBXIP up-regulates YAP through activation of transcription factor c-Myb to promote growth of liver cancer. Cancer Lett 2016; 385:234-242. [PMID: 27765671 DOI: 10.1016/j.canlet.2016.10.018] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Revised: 09/22/2016] [Accepted: 10/10/2016] [Indexed: 12/12/2022]
Abstract
The oncoprotein Yes-associated protein (YAP) in Hippo pathway plays crucial roles in the development of cancer. However, the mechanism of YAP regulation in cancer remains poorly understood. Here, we supposed that the oncoprotein hepatitis B X-interacting protein (HBXIP) might be involved in the modulation of YAP in liver cancer. Interestingly, our data showed that the expression levels of HBXIP were positively associated with those of YAP in clinical hepatocellular carcinoma (HCC) samples by immunohistochemistry (IHC) staining and real-time PCR assays. HBXIP was able to up-regulate YAP in hepatoma cells at the levels of promoter, mRNA and protein. Mechanistically, we identified that HBXIP up-regulated YAP through co-activating the transcription factor c-Myb in hepatoma cells. Functionally, silencing YAP abolished the proliferation of hepatoma cells mediated by HBXIP in vitro. Moreover, knockdown of YAP strongly blocked the HBXIP-enhanced tumor growth in mice. Thus, we conclude that HBXIP up-regulates YAP expression via activating transcription factor c-Myb to facilitate the growth of hepatoma cells. Our finding provides new insights into the mechanism of YAP regulation. Therapeutically, the oncoprotein HBXIP and YAP might serve as targets in liver cancer.
Collapse
Affiliation(s)
- Yue Wang
- State Key Laboratory of Medicinal Chemical Biology, Department of Biochemistry, College of Life Sciences, Nankai University, Tianjin, People's Republic of China
| | - Runping Fang
- State Key Laboratory of Medicinal Chemical Biology, Department of Biochemistry, College of Life Sciences, Nankai University, Tianjin, People's Republic of China
| | - Ming Cui
- State Key Laboratory of Medicinal Chemical Biology, Department of Cancer Research, College of Life Sciences, Nankai University, Tianjin, People's Republic of China
| | - Weiying Zhang
- State Key Laboratory of Medicinal Chemical Biology, Department of Biochemistry, College of Life Sciences, Nankai University, Tianjin, People's Republic of China
| | - Xiao Bai
- State Key Laboratory of Medicinal Chemical Biology, Department of Biochemistry, College of Life Sciences, Nankai University, Tianjin, People's Republic of China
| | - Huawei Wang
- State Key Laboratory of Medicinal Chemical Biology, Department of Biochemistry, College of Life Sciences, Nankai University, Tianjin, People's Republic of China
| | - Bowen Liu
- State Key Laboratory of Medicinal Chemical Biology, Department of Biochemistry, College of Life Sciences, Nankai University, Tianjin, People's Republic of China
| | - Xiaodong Zhang
- State Key Laboratory of Medicinal Chemical Biology, Department of Cancer Research, College of Life Sciences, Nankai University, Tianjin, People's Republic of China.
| | - Lihong Ye
- State Key Laboratory of Medicinal Chemical Biology, Department of Biochemistry, College of Life Sciences, Nankai University, Tianjin, People's Republic of China.
| |
Collapse
|
755
|
Horváth B, Betancourt AJ, Kalinka AT. A novel method for quantifying the rate of embryogenesis uncovers considerable genetic variation for the duration of embryonic development in Drosophila melanogaster. BMC Evol Biol 2016; 16:200. [PMID: 27717305 PMCID: PMC5054588 DOI: 10.1186/s12862-016-0776-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Accepted: 09/29/2016] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Embryogenesis is a highly conserved, canalized process, and variation in the duration of embryogenesis (DOE), i.e., time from egg lay to hatching, has a potentially profound effect on the outcome of within- and between-species competition. There is both intra- and inter-specific variation in this trait, which may provide important fuel for evolutionary processes, particularly adaptation. However, while genetic variation underlying simpler morphological traits, or with large phenotypic effects is well described in the literature, less is known about the underlying genetics of traits, such as DOE, partly due to a lack of tools with which to study them. RESULTS Here, we establish a novel microscope-based assay to survey genetic variation for the duration of embryogenesis (DOE). First, to establish the potential importance of DOE in competitive fitness, we performed a set of experiments where we experimentally manipulated the time until hatching, and show that short hatching times result in priority effect in the form of improved larval competitive ability. We then use our assay to measure DOE for 43 strains from the Drosophila Genetic Reference Panel (DGRP). Our assay greatly simplifies the measurement of DOE, making it possible to precisely quantify this trait for 59,295 individual embryos (mean ± S.D. of 1103 ± 293 per DGRP strain, and 1002 ± 203 per control). We find extensive genetic variation in DOE, with a 15 % difference in rate between the slowest and fastest strains measured, and 89 % of phenotypic variation due to DGRP strain. Using sequence information from the DGRP, we perform a genome-wide association study, which suggests that some well-known developmental genes affect the speed of embryonic development. CONCLUSIONS We showed that the duration of embryogenesis (DOE) can be efficiently and precisely measured in Drosophila, and that the DGRP strains show remarkable variation in DOE. A genome-wide analysis suggests that some well-known developmental genes are potentially associated with DOE. Further functional assays, or transcriptomic analysis of embryos from the DGRP, can validate the role of our candidates in early developmental processes.
Collapse
Affiliation(s)
- Barbara Horváth
- Institut für Populationsgenetik, Veterinärmedizinische Universität Wien, Veterinärplatz 1, A-1210, Vienna, Austria. .,Vienna Graduate School of Population Genetics, Veterinärmedizinische Universität Wien, Veterinärplatz 1, Vienna, A-1210, Austria.
| | - Andrea J Betancourt
- Institut für Populationsgenetik, Veterinärmedizinische Universität Wien, Veterinärplatz 1, A-1210, Vienna, Austria
| | - Alex T Kalinka
- Institut für Populationsgenetik, Veterinärmedizinische Universität Wien, Veterinärplatz 1, A-1210, Vienna, Austria
| |
Collapse
|
756
|
In Silico Analysis of Gene Expression Network Components Underlying Pigmentation Phenotypes in the Python Identified Evolutionarily Conserved Clusters of Transcription Factor Binding Sites. Adv Bioinformatics 2016; 2016:1286510. [PMID: 27698666 PMCID: PMC5028829 DOI: 10.1155/2016/1286510] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2015] [Revised: 03/17/2016] [Accepted: 06/02/2016] [Indexed: 12/22/2022] Open
Abstract
Color variation provides the opportunity to investigate the genetic basis of evolution and selection. Reptiles are less studied than mammals. Comparative genomics approaches allow for knowledge gained in one species to be leveraged for use in another species. We describe a comparative vertebrate analysis of conserved regulatory modules in pythons aimed at assessing bioinformatics evidence that transcription factors important in mammalian pigmentation phenotypes may also be important in python pigmentation phenotypes. We identified 23 python orthologs of mammalian genes associated with variation in coat color phenotypes for which we assessed the extent of pairwise protein sequence identity between pythons and mouse, dog, horse, cow, chicken, anole lizard, and garter snake. We next identified a set of melanocyte/pigment associated transcription factors (CREB, FOXD3, LEF-1, MITF, POU3F2, and USF-1) that exhibit relatively conserved sequence similarity within their DNA binding regions across species based on orthologous alignments across multiple species. Finally, we identified 27 evolutionarily conserved clusters of transcription factor binding sites within ~200-nucleotide intervals of the 1500-nucleotide upstream regions of AIM1, DCT, MC1R, MITF, MLANA, OA1, PMEL, RAB27A, and TYR from Python bivittatus. Our results provide insight into pigment phenotypes in pythons.
Collapse
|
757
|
Claro da Silva T, Hiller C, Gai Z, Kullak-Ublick GA. Vitamin D3 transactivates the zinc and manganese transporter SLC30A10 via the Vitamin D receptor. J Steroid Biochem Mol Biol 2016; 163:77-87. [PMID: 27107558 DOI: 10.1016/j.jsbmb.2016.04.006] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Revised: 03/15/2016] [Accepted: 04/13/2016] [Indexed: 01/11/2023]
Abstract
Vitamin D3 regulates genes critical for human health and its deficiency is associated with an increased risk for osteoporosis, cancer, diabetes, multiple sclerosis, hypertension, inflammatory and immunological diseases. To study the impact of vitamin D3 on genes relevant for the transport and metabolism of nutrients and drugs, we employed next-generation sequencing (NGS) and analyzed global gene expression of the human-derived Caco-2 cell line treated with 500nM vitamin D3. Genes involved in neuropeptide signaling, inflammation, cell adhesion and morphogenesis were differentially expressed. Notably, genes implicated in zinc, manganese and iron homeostasis were largely increased by vitamin D3 treatment. An ∼10-fold increase in ceruloplasmin and ∼4-fold increase in haptoglobin gene expression suggested a possible association between vitamin D and iron homeostasis. SLC30A10, the gene encoding the zinc and manganese transporter ZnT10, was the chiefly affected transporter, with ∼15-fold increase in expression. SLC30A10 is critical for zinc and manganese homeostasis and mutations in this gene, resulting in impaired ZnT10 function or expression, cause manganese intoxication, with Parkinson-like symptoms. Our NGS results were validated by real-time PCR in Caco-2 cells, as well as in duodenal biopsies taken from healthy human subjects treated with 0.5μg vitamin D3 daily for 10 days. In addition to increasing gene expression of SLC30A10 and the positive control TRPV6, vitamin D3 also increased ZnT10 protein expression, as indicated by Western blot and cytofluorescence. In silico identification of potential vitamin D responsive elements (VDREs) in the 5'-flanking region of the SLC30A10 promoter and dual-luciferase reporter assay showed enhanced promoter activity in the presence of vitamin D receptor (VDR) and retinoid X receptor (RXR) constructs, as well as vitamin D3, but not when one of these factors was absent. Electrophoretic mobility shift assay (EMSA) and competition EMSA revealed binding of select sequences, namely, nt -1623/-1588 and nt -1758/-1723 relative to the transcription start site, to VDR-containing nuclear extracts. In conclusion, we have shown that vitamin D3 transactivates the SLC30A10 gene in a VDR-dependent manner, resulting in increased ZnT10 protein expression. Because SLC30A10 is highly expressed in the small intestine, it is possible that the control of zinc and manganese systemic levels is regulated by vitamin D3 in the intestine. Zinc, manganese and vitamin D are important for bone metabolism and brain health. Future examination of a possible role for supplementation or chelation of zinc and manganese, alongside vitamin D3 administration, will further our understanding of its potential benefit in the treatment of specific illnesses, such as osteoporosis and Parkinson's disease.
Collapse
Affiliation(s)
- Tatiana Claro da Silva
- Department of Clinical Pharmacology and Toxicology, University Hospital Zurich, University of Zurich, Switzerland.
| | - Christian Hiller
- Department of Clinical Pharmacology and Toxicology, University Hospital Zurich, University of Zurich, Switzerland.
| | - Zhibo Gai
- Department of Clinical Pharmacology and Toxicology, University Hospital Zurich, University of Zurich, Switzerland.
| | - Gerd A Kullak-Ublick
- Department of Clinical Pharmacology and Toxicology, University Hospital Zurich, University of Zurich, Switzerland.
| |
Collapse
|
758
|
Chuang YA, Hu TM, Chen CH, Hsu SH, Tsai HY, Cheng MC. Rare mutations and hypermethylation of the ARC gene associated with schizophrenia. Schizophr Res 2016; 176:106-113. [PMID: 27464451 DOI: 10.1016/j.schres.2016.07.019] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Revised: 07/18/2016] [Accepted: 07/20/2016] [Indexed: 11/19/2022]
Abstract
Activity-regulated cytoskeleton-associated protein (ARC), which interacts with the N-methyl-d-aspartate receptor (NMDAR) complex, is a critical effector molecule downstream of multiple neuronal signaling pathways. Dysregulation of the ARC/NMDAR complex can disrupt learning, memory, and normal brain functions. This study examined the role of ARC in susceptibility to schizophrenia. We used a resequencing strategy to identify the variants of ARC in 1078 subjects, including patients with schizophrenia and normal controls. We identified 16 known SNPs and 27 rare mutations. SNP-based analysis showed no association of ARC with schizophrenia. In addition, the rare mutations did not increase the burden in patients compared with controls. However, one patient-specific allele in the putative ARC promoter region and seven patient-specific mutants in ARC exon regions significantly reduced the reporter gene activity compared with ARC wild-type. Methylation of a putative ARC promoter attenuated reporter activity in vitro, suggesting that ARC expression is regulated by DNA methylation. Pyrosequencing revealed eight hypermethylated CpG sites in the putative ARC promoter region in 64 schizophrenic patients compared with 63 controls. Taken together, our results suggest that both rare variants and epigenetic regulation of ARC contribute to the pathogenesis of schizophrenia in some patients.
Collapse
Affiliation(s)
- Yang-An Chuang
- Department of Psychiatry, Yuli Mental Health Research Center, Yuli Branch, Taipei Veterans General Hospital, Hualien, Taiwan
| | - Tsung-Ming Hu
- Department of Psychiatry, Yuli Mental Health Research Center, Yuli Branch, Taipei Veterans General Hospital, Hualien, Taiwan
| | - Chia-Hsiang Chen
- Department of Psychiatry, Chang Gung Memorial Hospital at Linkou and Department and Graduate School of Biomedical Sciences Chang Gung University, Taoyuan, Taiwan
| | - Shih-Hsin Hsu
- Department of Psychiatry, Yuli Mental Health Research Center, Yuli Branch, Taipei Veterans General Hospital, Hualien, Taiwan
| | - Hsin-Yao Tsai
- Department of Psychiatry, Yuli Mental Health Research Center, Yuli Branch, Taipei Veterans General Hospital, Hualien, Taiwan
| | - Min-Chih Cheng
- Department of Psychiatry, Yuli Mental Health Research Center, Yuli Branch, Taipei Veterans General Hospital, Hualien, Taiwan; Center for General Education, St. Mary's Junior College of Medicine, Nursing and Management, Yilan County, Taiwan.
| |
Collapse
|
759
|
Wang Y, Xu G, Han J, Xu T. miR-200a-3p regulates TLR1 expression in bacterial challenged miiuy croaker. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2016; 63:181-186. [PMID: 27288848 DOI: 10.1016/j.dci.2016.06.004] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Revised: 06/06/2016] [Accepted: 06/06/2016] [Indexed: 06/06/2023]
Abstract
MicroRNAs (miRNAs) are highly conserved, small non-coding RNAs which post-transcriptionally regulate various biological processes by repressing mRNA translation or degradating mRNA. It has been demonstrated that miRNAs play crucial roles in regulating the immune system. In this study, we explored the potential roles of miR-200a-3p in regulating TLR signaling pathway in miiuy croaker. Bioinformatics analysis showed that miiuy croaker TLR1 (mmiTLR1) was a putative target of miR-200a-3p. Negative expression profiles in spleen of Vibrio anguillarum challenged miiuy croaker and in lipopolysaccharide (LPS) stimulated miiuy croaker leukocytes further validated the prediction. Luciferase reporter assays showed that the dual-luciferase reporter fused to the 3'UTR of wild type mmiTLR1 cotransfected with miR-200a-3p mimics exhibited a reduction in luciferase activity compared with the controls. All of the present data provide direct evidence that miR-200a-3p is involved in TLR1 expression modulation in miiuy croaker, which will offer a basis for better understanding of miRNA regulation in fish TLR signaling pathways.
Collapse
Affiliation(s)
- Yanjin Wang
- Laboratory of Fish Biogenetics & Immune Evolution, College of Marine Science, Zhejiang Ocean University, Zhoushan, 316022, China
| | - Guoliang Xu
- Laboratory of Fish Biogenetics & Immune Evolution, College of Marine Science, Zhejiang Ocean University, Zhoushan, 316022, China
| | - Jingjing Han
- Laboratory of Fish Biogenetics & Immune Evolution, College of Marine Science, Zhejiang Ocean University, Zhoushan, 316022, China
| | - Tianjun Xu
- Laboratory of Fish Biogenetics & Immune Evolution, College of Marine Science, Zhejiang Ocean University, Zhoushan, 316022, China.
| |
Collapse
|
760
|
Zheng L, Jiao W, Song H, Qu H, Li D, Mei H, Chen Y, Yang F, Li H, Huang K, Tong Q. miRNA-558 promotes gastric cancer progression through attenuating Smad4-mediated repression of heparanase expression. Cell Death Dis 2016; 7:e2382. [PMID: 27685626 PMCID: PMC5059886 DOI: 10.1038/cddis.2016.293] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Revised: 08/20/2016] [Accepted: 08/23/2016] [Indexed: 12/25/2022]
Abstract
Previous studies have indicated that as the only mammalian endo-β-D-glucuronidase, heparanase (HPSE) is up-regulated and associated with poor prognosis in gastric cancer, while the underlying mechanisms still remain to be determined. Herein, through integrative analysis of public datasets, we found microRNA-558 (miR-558) and SMAD family member 4 (Smad4) as the crucial transcription regulators of HPSE expression in gastric cancer, with their adjacent target sites within the promoter of HPSE. We identified that endogenous miR-558 activated the transcription and expression of HPSE in gastric cancer cell lines. In contrast, Smad4 suppressed the nascent transcription and expression of HPSE via directly binding to its promoter. Mechanistically, miR-558 recognized its complementary site within HPSE promoter to decrease the binding of Smad4 in an Argonaute 1-dependent manner. Ectopic expression or knockdown experiments indicated that miR-558 promoted the in vitro and in vivo tumorigenesis and aggressiveness of gastric cancer cell lines via attenuating Smad4-mediated repression of HPSE expression. In clinical gastric cancer specimens, up-regulation of miR-558 and down-regulation of Smad4 were positively correlated with HPSE expression. Kaplan–Meier survival analysis revealed that miR-558 and Smad4 were associated with unfavourable and favourable outcome of gastric cancer patients, respectively. Therefore, these findings demonstrate that miR-558 facilitates the progression of gastric cancer through directly targeting the HPSE promoter to attenuate Smad4-mediated repression of HPSE expression.
Collapse
Affiliation(s)
- Liduan Zheng
- Department of Pathology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, Hubei Province, P. R. China.,Clinical Center of Human Genomic Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, Hubei Province, P. R. China
| | - Wanju Jiao
- Department of Pathology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, Hubei Province, P. R. China
| | - Huajie Song
- Department of Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, Hubei Province, P. R. China
| | - Hongxia Qu
- Department of Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, Hubei Province, P. R. China
| | - Dan Li
- Department of Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, Hubei Province, P. R. China
| | - Hong Mei
- Department of Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, Hubei Province, P. R. China
| | - Yajun Chen
- Department of Pathology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, Hubei Province, P. R. China
| | - Feng Yang
- Department of Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, Hubei Province, P. R. China
| | - Huanhuan Li
- Department of Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, Hubei Province, P. R. China
| | - Kai Huang
- Clinical Center of Human Genomic Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, Hubei Province, P. R. China
| | - Qiangsong Tong
- Clinical Center of Human Genomic Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, Hubei Province, P. R. China.,Department of Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, Hubei Province, P. R. China
| |
Collapse
|
761
|
PPARγ regulates exocrine pancreas lipase. Biochim Biophys Acta Mol Cell Biol Lipids 2016; 1861:1921-1928. [PMID: 27663184 DOI: 10.1016/j.bbalip.2016.09.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Revised: 09/12/2016] [Accepted: 09/15/2016] [Indexed: 12/30/2022]
Abstract
AIM Pancreatic lipase (triacylglycerol lipase EC 3.1.1.3) is an essential enzyme in hydrolysis of dietary fat. Dietary fat, especially polyunsaturated fatty acids (PUFA), regulate pancreatic lipase (PNLIP); however, the molecular mechanism underlying this regulation is mostly unknown. As PUFA are known to regulate expression of proliferator-activated receptor gamma (PPARγ), and as we identified in-silico putative PPARγ binding sites within the putative PNLIP promoter sequence, we hypothesized that PUFA regulation of PNLIP might be mediated by PPARγ. MATERIALS AND METHODS We used in silico bioinformatics tools, reporter luciferase assay, PPARγ agonists and antagonists, PPARγ overexpression in exocrine pancreas AR42J and primary cells to study PPARγ regulation of PNLIP. RESULTS Using in silico bioinformatics tools we mapped PPARγ binding sites (PPRE) to the putative promoter region of PNLIP. Reporter luciferase assay in AR42J rat exocrine pancreas acinar cells transfected with various constructs of the putative PNLIP promoter showed that PNLIP transcription is significantly enhanced by PPARγ dose-dependently, reaching maximal levels with multi PPRE sites. This effect was significantly augmented in the presence of PPARγ agonists and reduced by PPARγ antagonists or mutagenesis abrogating PPRE sites. Over-expression of PPARγ significantly elevated PNLIP transcript and protein levels in AR42J cells and in primary pancreas cells. Moreover, PNLIP expression was up-regulated by PPARγ agonists (pioglitazone and 15dPGJ2) and significantly down-regulated by PPARγ antagonists in non-transfected rat exocrine pancreas AR42J cell line cells. CONCLUSION PPARγ transcriptionally regulates PNLIP gene expression. This transcript regulation resolves part of the missing link between dietary PUFA direct regulation of PNLIP.
Collapse
|
762
|
Lopez-Bertoni H, Lal B, Michelson N, Guerrero-Cázares H, Quiñones-Hinojosa A, Li Y, Laterra J. Epigenetic modulation of a miR-296-5p:HMGA1 axis regulates Sox2 expression and glioblastoma stem cells. Oncogene 2016; 35:4903-13. [PMID: 26898758 PMCID: PMC6151872 DOI: 10.1038/onc.2016.22] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Revised: 12/17/2015] [Accepted: 12/18/2015] [Indexed: 12/15/2022]
Abstract
Solid malignancies contain subsets of multipotent cells that grow as spheres and efficiently propagate tumors in xenograft models, reflecting a stem-like, self-renewing and tumor-propagating phenotype. These cancer 'stem cells (SCs)' have been shown to maintain tumor growth, contribute to resistance and drive tumor recurrence. Cancer cell stemness is dynamically influenced by epigenetic mechanisms and differentially regulated coding and noncoding RNAs. How these mechanisms specifically contribute to the generation and/or maintenance of cancer SCs remains unclear. This study identifies a novel epigenetically regulated circuit that integrates microRNA, chromatin remodeling and the reprogramming transcription factor Sox2 to regulate glioblastoma (GBM)-propagating SCs. We show that miR-296-5p expression is repressed in a DNA methylation-dependent manner under conditions that promote GBM cell stemness and that miR-296-5p inhibits GBM cell stemness and their capacity to self-renew as spheres and propagate glioma xenografts in vivo. We show that the chromatin remodeling protein HMGA1 functions as a downstream effector of these biological responses to miR-296-5p and regulates Sox2 expression, a master driver of cell stemness, by modifying chromatin architecture at the Sox2 promoter. These results show for the first time that miR-296-5p inhibits transcriptional mechanisms that support GBM SCs and identify a miR-296-5p:HMGA1:Sox2 axis as a novel regulator of GBM SCs and candidate pathway for targeting therapies directed at depleting tumors of their tumor-propagating stem cell subsets.
Collapse
Affiliation(s)
- H Lopez-Bertoni
- Hugo W Moser Research Institute at Kennedy Krieger, Baltimore, MD, USA
- Department of Neurology, The Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - B Lal
- Hugo W Moser Research Institute at Kennedy Krieger, Baltimore, MD, USA
- Department of Neurology, The Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - N Michelson
- Hugo W Moser Research Institute at Kennedy Krieger, Baltimore, MD, USA
| | - H Guerrero-Cázares
- Department of Neurosurgery, The Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - A Quiñones-Hinojosa
- Department of Neurosurgery, The Johns Hopkins School of Medicine, Baltimore, MD, USA
- Department of Neuroscience, The Johns Hopkins School of Medicine, Baltimore, MD, USA
- Department of Oncology, The Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Y Li
- Hugo W Moser Research Institute at Kennedy Krieger, Baltimore, MD, USA
- Department of Neurology, The Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - J Laterra
- Hugo W Moser Research Institute at Kennedy Krieger, Baltimore, MD, USA
- Department of Neurology, The Johns Hopkins School of Medicine, Baltimore, MD, USA
- Department of Neuroscience, The Johns Hopkins School of Medicine, Baltimore, MD, USA
- Department of Oncology, The Johns Hopkins School of Medicine, Baltimore, MD, USA
| |
Collapse
|
763
|
Alcaíno J, Bravo N, Córdova P, Marcoleta AE, Contreras G, Barahona S, Sepúlveda D, Fernández-Lobato M, Baeza M, Cifuentes V. The Involvement of Mig1 from Xanthophyllomyces dendrorhous in Catabolic Repression: An Active Mechanism Contributing to the Regulation of Carotenoid Production. PLoS One 2016; 11:e0162838. [PMID: 27622474 PMCID: PMC5021340 DOI: 10.1371/journal.pone.0162838] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2016] [Accepted: 08/29/2016] [Indexed: 11/18/2022] Open
Abstract
The red yeast X. dendrorhous is one of the few natural sources of astaxanthin, a carotenoid used in aquaculture for salmonid fish pigmentation and in the cosmetic and pharmaceutical industries for its antioxidant properties. Genetic control of carotenogenesis is well characterized in this yeast; however, little is known about the regulation of the carotenogenesis process. Several lines of evidence have suggested that carotenogenesis is regulated by catabolic repression, and the aim of this work was to identify and functionally characterize the X. dendrorhous MIG1 gene encoding the catabolic repressor Mig1, which mediates transcriptional glucose-dependent repression in other yeasts and fungi. The identified gene encodes a protein of 863 amino acids that demonstrates the characteristic conserved features of Mig1 proteins, and binds in vitro to DNA fragments containing Mig1 boxes. Gene functionality was demonstrated by heterologous complementation in a S. cerevisiae mig1- strain; several aspects of catabolic repression were restored by the X. dendrorhous MIG1 gene. Additionally, a X. dendrorhous mig1- mutant was constructed and demonstrated a higher carotenoid content than the wild-type strain. Most important, the mig1- mutation alleviated the glucose-mediated repression of carotenogenesis in X. dendrorhous: the addition of glucose to mig1- and wild-type cultures promoted the growth of both strains, but carotenoid synthesis was observed only in the mutant strain. Transcriptomic and RT-qPCR analyses revealed that several genes were differentially expressed between X. dendrorhous mig1- and the wild-type strain when cultured with glucose as the sole carbon source. The results obtained in this study demonstrate that catabolic repression in X. dendrorhous is an active process in which the identified MIG1 gene product plays a central role in the regulation of several biological processes, including carotenogenesis.
Collapse
Affiliation(s)
- Jennifer Alcaíno
- Laboratorio de Genética, Departamento de Ciencias Ecológicas, Facultad de Ciencias, Universidad de Chile, Santiago, Chile
| | - Natalia Bravo
- Laboratorio de Genética, Departamento de Ciencias Ecológicas, Facultad de Ciencias, Universidad de Chile, Santiago, Chile
| | - Pamela Córdova
- Laboratorio de Genética, Departamento de Ciencias Ecológicas, Facultad de Ciencias, Universidad de Chile, Santiago, Chile
| | - Andrés E. Marcoleta
- Departamento de Biología, Facultad de Ciencias, Universidad de Chile, Santiago, Chile
| | - Gabriela Contreras
- Laboratorio de Genética, Departamento de Ciencias Ecológicas, Facultad de Ciencias, Universidad de Chile, Santiago, Chile
| | - Salvador Barahona
- Laboratorio de Genética, Departamento de Ciencias Ecológicas, Facultad de Ciencias, Universidad de Chile, Santiago, Chile
| | - Dionisia Sepúlveda
- Laboratorio de Genética, Departamento de Ciencias Ecológicas, Facultad de Ciencias, Universidad de Chile, Santiago, Chile
| | - María Fernández-Lobato
- Centro de Biología Molecular Severo Ochoa, Departamento de Biología Molecular (UAM-CSIC), Universidad Autónoma, Madrid, Cantoblanco, España
| | - Marcelo Baeza
- Laboratorio de Genética, Departamento de Ciencias Ecológicas, Facultad de Ciencias, Universidad de Chile, Santiago, Chile
| | - Víctor Cifuentes
- Laboratorio de Genética, Departamento de Ciencias Ecológicas, Facultad de Ciencias, Universidad de Chile, Santiago, Chile
- * E-mail:
| |
Collapse
|
764
|
The Promoter and Multiple Enhancers of the pou4f3 Gene Regulate Expression in Inner Ear Hair Cells. Mol Neurobiol 2016; 54:5414-5426. [PMID: 27592349 DOI: 10.1007/s12035-016-0060-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Accepted: 08/16/2016] [Indexed: 12/21/2022]
Abstract
Few enhancers that target gene expression to inner ear hair cells (HCs) have been identified. Using transgenic analysis of enhanced green fluorescent protein (eGFP) reporter constructs and bioinformatics, we evaluated the control of pou4f3 gene expression, since it is expressed only in HCs within the inner ear and continues to be expressed throughout life. An 8.5-kb genomic DNA fragment 5' to the start codon, containing three regions of high cross-species homology, drove expression in all embryonic and neonatal HCs, and adult vestibular and inner HCs, but not adult outer HCs. Transgenes with 0.4, 0.8, 2.5, or 6.5 kb of 5' DNA did not produce HC expression. However, addition of the region from 6.5 to 7.2 kb produced expression in vestibular HCs and neonatal basal turn outer HCs, which also implicated the region from 7.2 to 8.5 kb in inner and apical outer HC expression. Deletion of the region from 0.4 to 5.5 kb 5' from the 8.5-kb construct did not affect HC expression, further indicating lack of HC regulatory elements. When the region from 1 to 0.4 kb was replaced with the minimal promoter of the Ela1 gene, HC expression was maintained but at a drastically reduced level. Bioinformatics identified regions of highly conserved sequence outside of the 8.5 kb, which contained POU4F3-, GFI1-, and LHX3-binding sites. These regions may be involved in maintaining POU4F3 expression in adult outer HCs. Our results identify separate enhancers at various locations that direct expression to different HC types at different ages and determine that 0.4 kb of upstream sequence determines expression level. These data will assist in the identification of mutations in noncoding, regulatory regions of this deafness gene.
Collapse
|
765
|
Paolicchi E, Vivaldi C, De Gregorio V, Crea F, Fornaro L, Masi G, Loupakis F, Graziano F, Ronzoni M, Ricci V, Falcone A, Danesi R. Topoisomerase 1 Promoter Variants and Benefit from Irinotecan in Metastatic Colorectal Cancer Patients. Oncology 2016; 91:283-288. [PMID: 27577551 DOI: 10.1159/000448004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Accepted: 06/24/2016] [Indexed: 11/19/2022]
Abstract
OBJECTIVE Topoisomerase 1 (topo-1) is an important target for the treatment of metastatic colorectal cancer (CRC). The aim of the present study was to evaluate the correlation between topo-1 single-nucleotide polymorphisms (SNPs) and clinical outcome in metastatic CRC (mCRC) patients. METHODS With the use of specific software (PROMO 3.0), we performed an in silico analysis of topo-1 promoter SNPs; the rs6072249 and rs34282819 SNPs were included in the study. DNA was extracted from 105 mCRC patients treated with FOLFIRI ± bevacizumab in the first line. SNP genotyping was performed by real-time PCR. Genotypes were correlated with clinical parameters (objective response rate, progression-free survival, and overall survival). RESULTS No single genotype was significantly associated with clinical variables. The G allelic variant of rs6072249 topo-1 SNP is responsible for GC factor and X-box-binding protein transcription factor binding. The same allelic variant showed a nonsignificant trend toward a shorter progression-free survival (GG, 7.5 months; other genotypes, 9.3 months; HR 1.823, 95% CI 0.8904-3.734; p = 0.1). CONCLUSION Further analyses are needed to confirm that the topo-1 SNP rs6072249 and transcription factor interaction could be a part of tools to predict clinical outcome in mCRC patients treated with irinotecan-based regimens.
Collapse
Affiliation(s)
- Elisa Paolicchi
- Unit of Clinical Pharmacology and Pharmacogenetics, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
766
|
Fang X, Poulsen RR, Wang-Hu J, Shi O, Calvo NS, Simmons CS, Rivkees SA, Wendler CC. Knockdown of DNA methyltransferase 3a alters gene expression and inhibits function of embryonic cardiomyocytes. FASEB J 2016; 30:3238-55. [PMID: 27306334 PMCID: PMC5001511 DOI: 10.1096/fj.201600346r] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Accepted: 05/31/2016] [Indexed: 12/28/2022]
Abstract
We previously found that in utero caffeine exposure causes down-regulation of DNA methyltransferases (DNMTs) in embryonic heart and results in impaired cardiac function in adulthood. To assess the role of DNMTs in these events, we investigated the effects of reduced DNMT expression on embryonic cardiomyocytes. siRNAs were used to knock down individual DNMT expression in primary cultures of mouse embryonic cardiomyocytes. Immunofluorescence staining was conducted to evaluate cell morphology. A video-based imaging assay and multielectrode array were used to assess cardiomyocyte contractility and electrophysiology, respectively. RNA-Seq and multiplex bisulfite sequencing were performed to examine gene expression and promoter methylation, respectively. At 72 h after transfection, reduced DNMT3a expression, but not DNMT1 or -3b, disrupted sarcomere assembly and decreased beating frequency, contractile movement, amplitude of field action potential, and cytosolic calcium signaling of cardiomyocytes. RNA-Seq analysis revealed that the DNMT3a-deficient cells had deactivated gene networks involved in calcium, endothelin-1, renin-angiotensin, and cardiac β-adrenergic receptor signaling, which were not inhibited by DNMT3b siRNA. Moreover, decreased methylation levels were found in the promoters of Myh7, Myh7b, Tnni3, and Tnnt2, consistent with the up-regulation of these genes by DNMT3a siRNA. These data show that DNMT3a plays an important role in regulating embryonic cardiomyocyte gene expression, morphology and function.-Fang, X., Poulsen, R. R., Wang-Hu, J., Shi, O., Calvo, N. S., Simmons, C. S., Rivkees, S. A., Wendler, C. C. Knockdown of DNA methyltransferase 3a alters gene expression and inhibits function of embryonic cardiomyocytes.
Collapse
Affiliation(s)
- Xiefan Fang
- Child Health Research Institute, Department of Pediatrics, College of Medicine, University of Florida, Gainesville, Florida, USA; and
| | - Ryan R Poulsen
- Child Health Research Institute, Department of Pediatrics, College of Medicine, University of Florida, Gainesville, Florida, USA; and
| | - John Wang-Hu
- Child Health Research Institute, Department of Pediatrics, College of Medicine, University of Florida, Gainesville, Florida, USA; and
| | - Olivia Shi
- Child Health Research Institute, Department of Pediatrics, College of Medicine, University of Florida, Gainesville, Florida, USA; and
| | - Nicholas S Calvo
- Department of Mechanical and Aerospace Engineering, College of Engineering, University of Florida, Gainesville, Florida, USA
| | - Chelsey S Simmons
- Department of Mechanical and Aerospace Engineering, College of Engineering, University of Florida, Gainesville, Florida, USA
| | - Scott A Rivkees
- Child Health Research Institute, Department of Pediatrics, College of Medicine, University of Florida, Gainesville, Florida, USA; and
| | - Christopher C Wendler
- Child Health Research Institute, Department of Pediatrics, College of Medicine, University of Florida, Gainesville, Florida, USA; and
| |
Collapse
|
767
|
Hudler P, Britovsek NK, Grazio SF, Komel R. Association between polymorphisms in segregation genes BUB1B and TTK and gastric cancer risk. Radiol Oncol 2016; 50:297-307. [PMID: 27679546 PMCID: PMC5024654 DOI: 10.1515/raon-2015-0047] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Accepted: 08/09/2015] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Malignant transformation of normal gastric cells is a complex and multistep process, resulting in development of heterogeneous tumours. Susceptible genetic background, accumulation of genetic changes, and environmental factors play an important role in gastric carcinogenesis. Single nucleotide polymorphisms (SNPs) in mitotic segregation genes could be responsible for inducing the slow process of accumulation of genetic changes, leading to genome instability. PATIENTS AND METHODS We performed a case-control study of polymorphisms in mitotic kinases TTK rs151658 and BUB1B rs1031963 and rs1801376 to assess their effects on gastric cancer risk. We examined the TTK abundance in gastric cancer tissues using immunoblot analysis. RESULTS C/G genotype of rs151658 was more frequent in patients with diffuse type of gastric cancer and G/G genotype was more common in intestinal types of gastric cancers (p = 0.049). Polymorphic genotype A/A of rs1801376 was associated with higher risk for developing diffuse type of gastric cancer in female population (p = 0.007), whereas A/A frequencies were increased in male patients with subserosa tumour cell infiltration (p = 0.009). T/T genotype of rs1031963 was associated with well differentiated tumours (p = 0.035). TT+CT genotypes of rs1031963 and GG+AG genotypes of rs1801376 were significantly associated with gastric cancer risk (dominant model; OR = 2,929, 95% CI: 1.281-6.700; p = 0.017 and dominant model; OR = 0,364, 95% CI: 0.192-0.691; p = 0.003 respectively). CONCLUSIONS Our results suggest that polymorphisms in mitotic kinases TTK and BUB1B may contribute to gastric tumorigenesis and risk of tumour development. Further investigations on large populations and populations of different ethnicity are needed to determine their clinical utility.
Collapse
Affiliation(s)
- Petra Hudler
- University of Ljubljana, Faculty of Medicine, Institute of Biochemistry, Ljubljana, Slovenia
| | - Nina Kocevar Britovsek
- University of Ljubljana, Faculty of Medicine, Institute of Biochemistry, Ljubljana, Slovenia
| | - Snjezana Frkovic Grazio
- University Clinical Hospital Ljubljana, Department of Obstetrics and Gynecology, Department of Gynecological Pathology and Cytology, Ljubljana, Slovenia
| | - Radovan Komel
- University of Ljubljana, Faculty of Medicine, Institute of Biochemistry, Ljubljana, Slovenia
| |
Collapse
|
768
|
Contribution of polymorphic variation of inositol hexakisphosphate kinase 3 ( IP6K3 ) gene promoter to the susceptibility to late onset Alzheimer's disease. Biochim Biophys Acta Mol Basis Dis 2016; 1862:1766-73. [DOI: 10.1016/j.bbadis.2016.06.014] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Revised: 05/19/2016] [Accepted: 06/14/2016] [Indexed: 01/16/2023]
|
769
|
Rare Genetic Variants of the Transthyretin Gene Are Associated with Alzheimer's Disease in Han Chinese. Mol Neurobiol 2016; 54:5192-5200. [PMID: 27562180 DOI: 10.1007/s12035-016-0065-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Accepted: 08/17/2016] [Indexed: 02/02/2023]
Abstract
Alzheimer's disease (AD) is the most prevalent form of dementia in the world. The neuropathological characteristics of AD patients are the accumulation of extracellular plaques of β-amyloid (Aβ) and intracellular hyperphosphorylated tau protein. Transthyretin (TTR) may alleviate AD symptom by reducing Aβ concentration in the brain. There were reports for a decreased TTR level in both AD brain and blood. However, there is still no robust evidence to support the genetic association of the TTR gene with AD. In this study, we aimed to investigate the potential association of TTR variation with AD by directly sequencing the whole exons and the promoter region of the TTR gene in 529 AD patients and 334 healthy controls from Han Chinese population. We found no association between TTR common variants and AD but observed an enrichment of TTR rare variants in AD patients relative to controls. Further in silico prediction analysis and functional assessment at the cellular level identified four potentially pathogenic rare variants in AD patients. In particular, variant c.-239C>A could potentially downregulate the TTR promoter activity; c.200+4A>G might influence the constitutive splicing of TTR mRNA; c.148G>A (p.V50M) and c.332C>T (p.A111V) would change the structure of TTR and decrease its Aβ-binding ability. Our results provided direct genetic evidence to support the active involvement of TTR in AD.
Collapse
|
770
|
Farré M, Narayan J, Slavov GT, Damas J, Auvil L, Li C, Jarvis ED, Burt DW, Griffin DK, Larkin DM. Novel Insights into Chromosome Evolution in Birds, Archosaurs, and Reptiles. Genome Biol Evol 2016; 8:2442-51. [PMID: 27401172 PMCID: PMC5010900 DOI: 10.1093/gbe/evw166] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Homologous synteny blocks (HSBs) and evolutionary breakpoint regions (EBRs) in mammalian chromosomes are enriched for distinct DNA features, contributing to distinct phenotypes. To reveal HSB and EBR roles in avian evolution, we performed a sequence-based comparison of 21 avian and 5 outgroup species using recently sequenced genomes across the avian family tree and a newly-developed algorithm. We identified EBRs and HSBs in ancestral bird, archosaurian (bird, crocodile, and dinosaur), and reptile chromosomes. Genes involved in the regulation of gene expression and biosynthetic processes were preferably located in HSBs, including for example, avian-specific HSBs enriched for genes involved in limb development. Within birds, some lineage-specific EBRs rearranged genes were related to distinct phenotypes, such as forebrain development in parrots. Our findings provide novel evolutionary insights into genome evolution in birds, particularly on how chromosome rearrangements likely contributed to the formation of novel phenotypes.
Collapse
Affiliation(s)
- Marta Farré
- Department of Comparative Biomedical Sciences, Royal Veterinary College, Royal College Street, University of London, NW1 0TU, UK
| | - Jitendra Narayan
- Institute of Biological, Environmental and Rural Sciences, Aberystwyth University, SY23 3DA, UK
| | - Gancho T Slavov
- Institute of Biological, Environmental and Rural Sciences, Aberystwyth University, SY23 3DA, UK
| | - Joana Damas
- Department of Comparative Biomedical Sciences, Royal Veterinary College, Royal College Street, University of London, NW1 0TU, UK
| | - Loretta Auvil
- Illinois Informatics Institute, University of Illinois, Urbana, IL 61801, USA
| | - Cai Li
- China National GeneBank, BGI-Shenzhen, Shenzhen 518083, China Centre for GeoGenetics, Natural History Museum of Denmark, University of Copenhagen, Copenhagen, 1350, Denmark
| | - Erich D Jarvis
- Department of Neurobiology, Duke University Medical Center, Durham, NC, 27710, USA Howard Hughes Medical Institute, Chevy Chase, MD, 20815, USA
| | - David W Burt
- Department of Genomics and Genetics, the Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Midlothian EH25 9RG, UK
| | - Darren K Griffin
- School of Biosciences, University of Kent, Canterbury CT2 7NJ, UK
| | - Denis M Larkin
- Department of Comparative Biomedical Sciences, Royal Veterinary College, Royal College Street, University of London, NW1 0TU, UK
| |
Collapse
|
771
|
Vargas-Pinilla P, Babb P, Nunes L, Paré P, Rosa G, Felkl A, Longo D, Salzano FM, Paixão-Côrtes VR, Gonçalves GL, Bortolini MC. Progesterone Response Element Variation in the OXTR Promoter Region and Paternal Care in New World Monkeys. Behav Genet 2016; 47:77-87. [DOI: 10.1007/s10519-016-9806-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2015] [Accepted: 08/10/2016] [Indexed: 01/25/2023]
|
772
|
Patterson JR, Kim EJ, Goudreau JL, Lookingland KJ. FosB and ΔFosB expression in brain regions containing differentially susceptible dopamine neurons following acute neurotoxicant exposure. Brain Res 2016; 1649:53-66. [PMID: 27566062 DOI: 10.1016/j.brainres.2016.08.030] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Revised: 08/20/2016] [Accepted: 08/22/2016] [Indexed: 11/29/2022]
Abstract
Parkinson disease (PD) is characterized by progressive neuronal degeneration, in particular nigrostriatal dopamine (NSDA) neurons and consequent deficits in movement. In mice and non-human primates, NSDA neurons preferentially degenerate following exposure to the neurotoxicant 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP). Tuberoinfundibular (TI) DA neurons, in contrast, appear to be unaffected in PD and recover following acute MPTP exposure-induced injury (Behrouz et al., 2007; Benskey et al., 2012). The recovery of the TIDA neurons is dependent on de novo protein synthesis and positively correlated with an increase in parkin mRNA and protein expression (Benskey et al., 2012, 2015). Inhibition of parkin upregulation renders TIDA neurons susceptible to degeneration following MPTP exposure. In addition to parkin, other potentially protective proteins are likely to be differentially regulated in TIDA and NSDA neurons following neurotoxicant exposure. The regulation of potential transcription factors for parkin and other neuroprotective pathway genes are of interest since they may provide novel targets for PD disease modifying therapies. As such, we sought to determine if there are time-dependent differences in the expression of AP-1 transcription factors c-Fos, c-Jun, FosB, ΔFosB and JunD in TIDA and NSDA neurons of mice following acute MPTP exposure. We observed that both FosB and ΔFosB expression increase in brain regions containing TIDA, but not NSDA neurons. Furthermore, the nuclear and long-term expression of ΔFosB is consistent with its role as a transcription factor that may influence parkin transcription, which may underlie the unique ability of TIDA neurons to recovery from an injury that leads NSDA neurons to degeneration.
Collapse
Affiliation(s)
| | - Elizabeth J Kim
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, USA
| | - John L Goudreau
- Genetics Program, Michigan State University, East Lansing, MI, USA; Department of Neurology and Ophthalmology, Michigan State University, East Lansing, MI, USA; Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, USA; Neuroscience Program, Michigan State University, East Lansing, MI, USA; College of Osteopathic Medicine, Michigan State University, East Lansing, MI, USA
| | - Keith J Lookingland
- Genetics Program, Michigan State University, East Lansing, MI, USA; Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, USA; Neuroscience Program, Michigan State University, East Lansing, MI, USA; College of Osteopathic Medicine, Michigan State University, East Lansing, MI, USA.
| |
Collapse
|
773
|
Chen Y, Sun Y, Rao Q, Xu H, Li L, Chang C. Androgen receptor (AR) suppresses miRNA-145 to promote renal cell carcinoma (RCC) progression independent of VHL status. Oncotarget 2016; 6:31203-15. [PMID: 26304926 PMCID: PMC4741598 DOI: 10.18632/oncotarget.4522] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2014] [Accepted: 04/23/2015] [Indexed: 12/18/2022] Open
Abstract
Mutational inactivation of the VHL tumor suppressor plays key roles in the development of renal cell carcinoma (RCC), and mutated VHL-mediated VEGF induction has become the main target for the current RCC therapy. Here we identified a signal pathway of VEGF induction by androgen receptor (AR)/miRNA-145 as a new target to suppress RCC progression. Mechanism dissection revealed that AR might function through binding to the androgen receptor element (ARE) located on the promoter region of miRNA-145 to suppress p53's ability to induce expression of miRNA-145 that normally suppresses expression of HIF2α/VEGF/MMP9/CCND1. Suppressing AR with AR-shRNA or introducing exogenous miRNA-145 mimic can attenuate RCC progression independent of VHL status. MiR-145 mimic in preclinical RCC orthotopic xenograft mouse model revealed its efficacy in suppression of RCC progression. These results together identified signals by AR-suppressed miRNA-145 as a key player in the RCC progression via regulating HIF2α/VEGF/MMP9/CCND1 expression levels. Blockade of the newly identified signal by AR inhibition or miRNA-145 mimics has promising therapeutic benefit to suppress RCC progression.
Collapse
Affiliation(s)
- Yuan Chen
- Sex Hormone Research Center, Department of Urology, Tongji Medical College/Hospital, Huazhong University of Science and Technology, Wuhan, China.,George Whipple Lab for Cancer Research, Departments of Pathology, Urology, and Radiation Oncology and Wilmot Cancer Center, University of Rochester Medical Center, Rochester, NY, USA
| | - Yin Sun
- George Whipple Lab for Cancer Research, Departments of Pathology, Urology, and Radiation Oncology and Wilmot Cancer Center, University of Rochester Medical Center, Rochester, NY, USA
| | - Qun Rao
- Department of Gynaecology and Obstetrics, Tongji Medical College/Hospital, Huazhong University of Science and Technology, Wuhan, China.,George Whipple Lab for Cancer Research, Departments of Pathology, Urology, and Radiation Oncology and Wilmot Cancer Center, University of Rochester Medical Center, Rochester, NY, USA
| | - Hua Xu
- George Whipple Lab for Cancer Research, Departments of Pathology, Urology, and Radiation Oncology and Wilmot Cancer Center, University of Rochester Medical Center, Rochester, NY, USA
| | - Lei Li
- George Whipple Lab for Cancer Research, Departments of Pathology, Urology, and Radiation Oncology and Wilmot Cancer Center, University of Rochester Medical Center, Rochester, NY, USA
| | - Chawnshang Chang
- George Whipple Lab for Cancer Research, Departments of Pathology, Urology, and Radiation Oncology and Wilmot Cancer Center, University of Rochester Medical Center, Rochester, NY, USA.,Sex Hormone Research Center, China Medical University/Hospital, Taichung, Taiwan
| |
Collapse
|
774
|
Mardahl M, Schröter MF, Engelbert D, Pink M, Sperandio M, Hamann A, Syrbe U. Core 2 ß1,6-N-acetylglucosaminyltransferase-I, crucial for P-selectin ligand expression is controlled by a distal enhancer regulated by STAT4 and T-bet in CD4+ T helper cells 1. Mol Immunol 2016; 77:132-40. [PMID: 27505708 DOI: 10.1016/j.molimm.2016.08.001] [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: 04/04/2016] [Revised: 07/23/2016] [Accepted: 08/02/2016] [Indexed: 01/02/2023]
Abstract
P-selectin ligands (P-ligs) support the recruitment of lymphocytes into inflamed tissues. Binding to P-selectin is mediated by oligosaccharide groups synthesized by means of several glycosyltransferases including core 2 ß1,6-N-acetylglucosaminyltransferase-I (C2GlcNAcT-I), encoded by the gene Gcnt1. Using Gcnt1(-/-) Th1 cells, we show that C2GlcNAcT-I is crucial for inflammatory T cell homing in vivo. To understand the molecular regulation of Gcnt1 in CD4(+) T helper cells, we performed ChIP-on-chip experiments across the Gcnt1 locus assessing the chromatin structure in P-lig-expressing versus non-expressing CD4(+) T cells. This identified a distal region about 20kb upstream of the promoter where the presence of a H3K27me3 mark correlated with Gcnt1 repression. This region possessed IL-12-dependent enhancer activity in reporter assays, in accordance with preferential IL-12-dependent induction of Gcnt1 in vitro. STAT4 and T-bet cooperated in control of the enhancer activity. Deficiency in either one resulted in drastically reduced Gcnt1 mRNA expression in differentiated Th1 cells. While both STAT4 and T-bet were bound to the enhancer early after activation only T-bet binding persisted throughout the expansion phase after TCR signal cessation. This suggests sequential action of STAT4 and T-bet at the enhancer. In summary, we show that Gcnt1 transcription and subsequent P-lig induction in Th1 cells is governed by binding of STAT4 and T-bet to a distal enhancer and further regulated by epigenetic marks such as H3K27me3.
Collapse
Affiliation(s)
- Maibritt Mardahl
- Deutsches Rheumaforschungszentrum, Experimentelle Rheumatologie, Berlin, Germany
| | - Micha F Schröter
- Deutsches Rheumaforschungszentrum, Experimentelle Rheumatologie, Berlin, Germany
| | - Dirk Engelbert
- Deutsches Rheumaforschungszentrum, Experimentelle Rheumatologie, Berlin, Germany
| | - Matthias Pink
- Deutsches Rheumaforschungszentrum, Experimentelle Rheumatologie, Berlin, Germany
| | - Markus Sperandio
- Walter Brendel Zentrum für Experimentelle Medizin, Ludwig-Maximilians-Universität, Munich, Germany
| | - Alf Hamann
- Deutsches Rheumaforschungszentrum, Experimentelle Rheumatologie, Berlin, Germany
| | - Uta Syrbe
- Deutsches Rheumaforschungszentrum, Experimentelle Rheumatologie, Berlin, Germany; Charité Universitätsmedizin Berlin, Medizinische Klinik für Gastroenterologie, Infektiologie und Rheumatologie, Hindenburgdamm 30, 12200 Berlin, Germany.
| |
Collapse
|
775
|
Lo CL, Lossie AC, Liang T, Liu Y, Xuei X, Lumeng L, Zhou FC, Muir WM. High Resolution Genomic Scans Reveal Genetic Architecture Controlling Alcohol Preference in Bidirectionally Selected Rat Model. PLoS Genet 2016; 12:e1006178. [PMID: 27490364 PMCID: PMC4973992 DOI: 10.1371/journal.pgen.1006178] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Accepted: 06/15/2016] [Indexed: 12/30/2022] Open
Abstract
Investigations on the influence of nature vs. nurture on Alcoholism (Alcohol Use Disorder) in human have yet to provide a clear view on potential genomic etiologies. To address this issue, we sequenced a replicated animal model system bidirectionally-selected for alcohol preference (AP). This model is uniquely suited to map genetic effects with high reproducibility, and resolution. The origin of the rat lines (an 8-way cross) resulted in small haplotype blocks (HB) with a corresponding high level of resolution. We sequenced DNAs from 40 samples (10 per line of each replicate) to determine allele frequencies and HB. We achieved ~46X coverage per line and replicate. Excessive differentiation in the genomic architecture between lines, across replicates, termed signatures of selection (SS), were classified according to gene and region. We identified SS in 930 genes associated with AP. The majority (50%) of the SS were confined to single gene regions, the greatest numbers of which were in promoters (284) and intronic regions (169) with the least in exon's (4), suggesting that differences in AP were primarily due to alterations in regulatory regions. We confirmed previously identified genes and found many new genes associated with AP. Of those newly identified genes, several demonstrated neuronal function involved in synaptic memory and reward behavior, e.g. ion channels (Kcnf1, Kcnn3, Scn5a), excitatory receptors (Grin2a, Gria3, Grip1), neurotransmitters (Pomc), and synapses (Snap29). This study not only reveals the polygenic architecture of AP, but also emphasizes the importance of regulatory elements, consistent with other complex traits. Alcohol Used Disorder (AUD) or Alcoholism extracts a great societal cost in terms of human suffering. Understanding the genetic basis is critical to comprehend, treat and prevent this disease, but difficult in humans, as choice is influenced by nature and nurture. To discover its genetic basis, we used an animal model system that controlled for genetic and non-genetic factors through randomization, study replication, long-term divergent selection, and a controlled environment. We conducted whole genome sequencing in breeds that were either compulsive excessive drinkers or completely abstinent. We discovered consistent alterations in several genes and neurological pathways previously unassociated with alcoholism. These results strengthened our understanding of the genetic basis of alcoholism and revealed potential genetic- and neurological-based treatments.
Collapse
Affiliation(s)
- Chiao-Ling Lo
- Indiana Alcohol Research Center, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
- Department of Anatomy & Cell Biology, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
| | - Amy C. Lossie
- Indiana Alcohol Research Center, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
- Department of Animal Sciences, Purdue University, West Lafayette, Indiana, United States of America
| | - Tiebing Liang
- Indiana Alcohol Research Center, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
- Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
| | - Yunlong Liu
- Indiana Alcohol Research Center, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
- Department of Medical Genetics, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
| | - Xiaoling Xuei
- Indiana Alcohol Research Center, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
- Department of Biochemistry, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
| | - Lawrence Lumeng
- Indiana Alcohol Research Center, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
- Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
| | - Feng C. Zhou
- Indiana Alcohol Research Center, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
- Department of Anatomy & Cell Biology, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
- Stark Neuroscience Research Institute, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
- * E-mail: (FCZ); (WMM)
| | - William M. Muir
- Indiana Alcohol Research Center, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
- Department of Animal Sciences, Purdue University, West Lafayette, Indiana, United States of America
- Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
- * E-mail: (FCZ); (WMM)
| |
Collapse
|
776
|
Pinceti E, Shults CL, Rao YS, Pak TR. Differential Effects of E2 on MAPK Activity in the Brain and Heart of Aged Female Rats. PLoS One 2016; 11:e0160276. [PMID: 27487271 PMCID: PMC4972350 DOI: 10.1371/journal.pone.0160276] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Accepted: 07/15/2016] [Indexed: 12/21/2022] Open
Abstract
Aging and the coincident loss of circulating estrogens at menopause lead to increased risks for neurological and cardiovascular pathologies. Clinical studies show that estrogen therapy (ET) can be beneficial in mitigating these negative effects, in both the brain and heart, when it is initiated shortly after the perimenopausal transition. However, this same therapy is detrimental when initiated >10 years postmenopause. Importantly, the molecular mechanisms underlying this age-related switch in ET efficacy are unknown. Estrogen receptors (ERs) mediate the neuroprotective and cardioprotective functions of estrogens by modulating gene transcription or, non-genomically, by activating second messenger signaling pathways, such as mitogen activated protein kinases (MAPK). These kinases are critical regulators of cell signaling pathways and have widespread downstream effects. Our hypothesis is that age and estrogen deprivation following menopause alters the expression and activation of the MAPK family members p38 and ERK in the brain and heart. To test this hypothesis, we used a surgically induced model of menopause in 18 month old rats through bilateral ovariectomy (OVX) followed by an acute dose of 17β-estradiol (E2) administered at varying time points post-OVX (1 week, 4 weeks, 8 weeks, or 12 weeks). Age and E2 treatment differentially regulated kinase activity in both the brain and heart, and the effects were also brain region specific. MAPK signaling plays an integral role in aging, and the aberrant regulation of those signaling pathways might be involved in age-related disorders. Clinical studies show benefits of ET during early menopause but detrimental effects later, which might be reflective of changes in kinase expression and activation status.
Collapse
Affiliation(s)
- Elena Pinceti
- Department of Cell and Molecular Physiology, Health Science Division, Loyola University Chicago, Maywood, Illinois, United States of America
| | - Cody L. Shults
- Department of Cell and Molecular Physiology, Health Science Division, Loyola University Chicago, Maywood, Illinois, United States of America
| | - Yathindar S. Rao
- Department of Cell and Molecular Physiology, Health Science Division, Loyola University Chicago, Maywood, Illinois, United States of America
| | - Toni R. Pak
- Department of Cell and Molecular Physiology, Health Science Division, Loyola University Chicago, Maywood, Illinois, United States of America
- * E-mail:
| |
Collapse
|
777
|
Yoon HK, An HK, Ko MJ, Kim KS, Mun SW, Kim DH, Kim CM, Kim CH, Choi YW, Lee YC. Upregulation of Human ST8Sia VI (α2,8-Sialyltransferase) Gene Expression by Physcion in SK-N-BE(2)-C Human Neuroblastoma Cells. Int J Mol Sci 2016; 17:1246. [PMID: 27490539 PMCID: PMC5000644 DOI: 10.3390/ijms17081246] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Revised: 06/28/2016] [Accepted: 07/25/2016] [Indexed: 12/12/2022] Open
Abstract
In this research, we firstly demonstrated that physcion, an anthraquinone derivative, specifically increased the expression of the human α2,8-sialyltransferase (hST8Sia VI) gene in SK-N-BE(2)-C human neuroblastoma cells. To establish the mechanism responsible for the up-regulation of hST8Sia VI gene expression in physcion-treated SK-N-BE(2)-C cells, the putative promoter region of the hST8Sia VI gene was functionally characterized. Promoter analysis with serially truncated fragments of the 5'-flanking region showed that the region between -320 and -240 is crucial for physcion-induced transcription of hST8Sia VI in SK-N-BE(2)-C cells. Putative binding sites for transcription factors Pax-5 and NF-Y are located at this region. The Pax-5 binding site at -262 to -256 was essential for the expression of the hST8Sia VI gene by physcion in SK-N-BE(2)-C cells. Moreover, the transcription of hST8Sia VI induced by physcion in SK-N-BE(2)-C cells was inhibited by extracellular signal-regulated protein kinase (ERK) inhibitor U0126 and p38 mitogen-activated protein kinase (MAPK) inhibitor SB203580, but not c-Jun N-terminal kinase (JNK) inhibitor SP600125. These results suggest that physcion upregulates hST8Sia VI gene expression via ERK and p38 MAPK pathways in SK-N-BE(2)-C cells.
Collapse
Affiliation(s)
- Hyun-Kyoung Yoon
- Department of Medicinal Biotechnology, College of Health Sciences, Dong-A university, Busan 49315, Korea.
| | - Hyun-Kyu An
- Department of Medicinal Biotechnology, College of Health Sciences, Dong-A university, Busan 49315, Korea.
| | - Min Jung Ko
- Department of Horticultural Bioscience, Pusan National University, Miryang 50463, Korea.
| | - Kyoung-Sook Kim
- Department of Medicinal Biotechnology, College of Health Sciences, Dong-A university, Busan 49315, Korea.
| | - Seo-Won Mun
- Department of Medicinal Biotechnology, College of Health Sciences, Dong-A university, Busan 49315, Korea.
| | - Dong-Hyun Kim
- Department of Medicinal Biotechnology, College of Health Sciences, Dong-A university, Busan 49315, Korea.
| | - Cheol Min Kim
- Research Center for Anti-Aging Technology Development, Pusan National University, Busan 46241, Korea.
| | - Cheorl-Ho Kim
- Molecular and Cellular Glycobiology Unit, Department of Biological Sciences, SungKyunKwan University, Kyunggi-Do 16419, Korea.
| | - Young Whan Choi
- Department of Horticultural Bioscience, Pusan National University, Miryang 50463, Korea.
| | - Young-Choon Lee
- Department of Medicinal Biotechnology, College of Health Sciences, Dong-A university, Busan 49315, Korea.
| |
Collapse
|
778
|
Tiscia GL, Dørum E, Myklebust CF, Grandone E, Sandset PM, Skretting G. Functional characterization of annexin A5 gene promoter allelic variants. Thromb Res 2016; 144:93-9. [DOI: 10.1016/j.thromres.2016.06.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Revised: 05/23/2016] [Accepted: 06/12/2016] [Indexed: 10/21/2022]
|
779
|
The MicroRNA 29 Family Promotes Type II Cell Differentiation in Developing Lung. Mol Cell Biol 2016; 36:2141. [PMID: 27215389 DOI: 10.1128/mcb.00096-16] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Accepted: 05/16/2016] [Indexed: 12/16/2022] Open
Abstract
Lung alveolar type II cells uniquely synthesize surfactant, a developmentally regulated lipoprotein that is essential for breathing. Expression of the gene (SFTPA) encoding the major surfactant protein, SP-A, in midgestation human fetal lung (HFL) is dramatically induced by cyclic AMP (cAMP). cAMP induction of SP-A expression is repressed by transforming growth factor β (TGF-β) and by hypoxia. In this study, we found that expression of the microRNA 29 (miR-29) family was significantly upregulated in epithelial cells isolated from mouse fetal lung during late gestation and in epithelial cells isolated from HFL explants during type II cell differentiation in culture. miR-29 expression in cultured HFL epithelial cells was increased by cAMP and inhibited by hypoxia, whereas the miR-29 target, TGF-β2, was coordinately decreased. Knockdown of the miR-29 family in cultured HFL type II cells blocked cAMP-induced SP-A expression and accumulation of surfactant-containing lamellar bodies, suggesting their physiological relevance. This occurred through derepression of TGF-β signaling. Notably, cAMP increased binding of endogenous thyroid transcription factor 1 (TTF-1/Nkx2.1) to the miR-29ab1 promoter in HFL type II cells, and TTF-1 increased miR-29ab1 promoter-driven luciferase activity in cotransfection assays. Together, these findings identify miR-29 family members as TTF-1-driven mediators of SP-A expression and type II cell differentiation through repression of TGF-β signaling.
Collapse
|
780
|
Carraro JCC, Hermsdorff HHM, Mansego ML, Zulet MÁ, Milagro FI, Bressan J, Martínez JA. Higher Fruit Intake Is Related to TNF-α Hypomethylation and Better Glucose Tolerance in Healthy Subjects. JOURNAL OF NUTRIGENETICS AND NUTRIGENOMICS 2016; 9:95-105. [PMID: 27467584 DOI: 10.1159/000448101] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Accepted: 06/30/2016] [Indexed: 11/19/2022]
Abstract
BACKGROUND/AIM This study hypothesized an association between healthy dietary patterns, hypermethylation of the tumor necrosis factor-α (TNF-α) promoter and decreased risk of metabolic changes. METHODS Forty normal-weight young women were involved in this cross-sectional study. DNA was isolated from white blood cells, and CpG site methylation in TNF-α was analyzed by Sequenom EpiTyper. The quality of the diet was assessed by Healthy Eating Index (HEI-2005). RESULTS Contradicting our hypothesis, HEI-2005 score was negatively associated with CpG5 (r = -0.460, p = 0.003) and TNF-α total methylation (r = -0.355, p = 0.026). A higher intake of fruits was related to lower insulin, HOMA-IR, and TNF-α methylation. No other dietary pattern was related to TNF-α methylation. TNF-α total methylation correlated positively with systolic blood pressure (r = 0.323; p = 0.042) and CpG5 methylation with body mass index (r = 0.333, p = 0.036). Furthermore, fiber intake was negatively associated with the CpG5 (r = -0.324, p = 0.041) and TNF-α total methylation (r = -0.434, p = 0.005), whereas vitamin C intake was negatively associated with TNF-α total methylation (r = -0.411, p = 0.009). Intakes of apples and citrus fruits were negatively associated with TNF-α total methylation. CONCLUSION A healthy dietary pattern and higher fruit intake (particularly apples and citrus fruits) were related to better glucose tolerance in healthy subjects, which could be mediated by lower TNF-α methylation.
Collapse
|
781
|
Ghosh T, Barik S, Bhuniya A, Dhar J, Dasgupta S, Ghosh S, Sarkar M, Guha I, Sarkar K, Chakrabarti P, Saha B, Storkus WJ, Baral R, Bose A. Tumor-associated mesenchymal stem cells inhibit naïve T cell expansion by blocking cysteine export from dendritic cells. Int J Cancer 2016; 139:2068-81. [PMID: 27405489 DOI: 10.1002/ijc.30265] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Revised: 06/08/2016] [Accepted: 06/29/2016] [Indexed: 12/28/2022]
Abstract
Mesenchymal stem cells (MSCs) represent an important cellular constituent of the tumor microenvironment, which along with tumor cells themselves, serve to regulate protective immune responses in support of progressive disease. We report that tumor MSCs prevent the ability of dendritic cells (DC) to promote naïve CD4(+) and CD8(+) T cell expansion, interferon gamma secretion and cytotoxicity against tumor cells, which are critical to immune-mediated tumor eradication. Notably, tumor MSCs fail to prevent DC-mediated early T cell activation events or the ability of responder T cells to produce IL-2. The immunoregulatory activity of tumor MSCs is IL-10- and STAT3-dependent, with STAT3 repressing DC expression of cystathionase, a critical enzyme that converts methionine-to-cysteine. Under cysteine-deficient priming conditions, naïve T cells exhibit defective cellular metabolism and proliferation. Bioinformatics analyses as well as in vitro observations suggest that STAT3 may directly bind to a GAS-like motif within the cystathionase promoter (-269 to -261) leading to IL-10-STAT3 mediated repression of cystathionase gene transcription. Our collective results provide evidence for a novel mechanism of tumor MSC-mediated T cell inhibition within tumor microenvironment.
Collapse
Affiliation(s)
- Tithi Ghosh
- Department of Immunoregulation and Immunodiagnostics, Chittaranjan National Cancer Institute (CNCI), Kolkata, West Bengal, 700026, India
| | - Subhasis Barik
- Department of Immunoregulation and Immunodiagnostics, Chittaranjan National Cancer Institute (CNCI), Kolkata, West Bengal, 700026, India
| | - Avishek Bhuniya
- Department of Immunoregulation and Immunodiagnostics, Chittaranjan National Cancer Institute (CNCI), Kolkata, West Bengal, 700026, India
| | - Jesmita Dhar
- Bioinformatics Centre and Department of Biochemistry, Bose Institute, Kolkata, West Bengal, 700054, India
| | - Shayani Dasgupta
- Department of Immunoregulation and Immunodiagnostics, Chittaranjan National Cancer Institute (CNCI), Kolkata, West Bengal, 700026, India
| | - Sarbari Ghosh
- Department of Immunoregulation and Immunodiagnostics, Chittaranjan National Cancer Institute (CNCI), Kolkata, West Bengal, 700026, India
| | - Madhurima Sarkar
- Department of Immunoregulation and Immunodiagnostics, Chittaranjan National Cancer Institute (CNCI), Kolkata, West Bengal, 700026, India
| | - Ipsita Guha
- Department of Immunoregulation and Immunodiagnostics, Chittaranjan National Cancer Institute (CNCI), Kolkata, West Bengal, 700026, India
| | - Koustav Sarkar
- SRM Research Institute and Department of Biotechnology, SRM University, Chennai, Tamil Nadu, 603203, India
| | - Pinak Chakrabarti
- Bioinformatics Centre and Department of Biochemistry, Bose Institute, Kolkata, West Bengal, 700054, India
| | - Bhaskar Saha
- National Centre for Cell Science, Pune, Maharashtra, 411007, India
| | - Walter J Storkus
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Rathindranath Baral
- Department of Immunoregulation and Immunodiagnostics, Chittaranjan National Cancer Institute (CNCI), Kolkata, West Bengal, 700026, India
| | - Anamika Bose
- Department of Immunoregulation and Immunodiagnostics, Chittaranjan National Cancer Institute (CNCI), Kolkata, West Bengal, 700026, India
| |
Collapse
|
782
|
Day SE, Coletta RL, Kim JY, Campbell LE, Benjamin TR, Roust LR, De Filippis EA, Dinu V, Shaibi GQ, Mandarino LJ, Coletta DK. Next-generation sequencing methylation profiling of subjects with obesity identifies novel gene changes. Clin Epigenetics 2016; 8:77. [PMID: 27437034 PMCID: PMC4950754 DOI: 10.1186/s13148-016-0246-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Accepted: 07/11/2016] [Indexed: 01/06/2023] Open
Abstract
Background Obesity is a metabolic disease caused by environmental and genetic factors. However, the epigenetic mechanisms of obesity are incompletely understood. The aim of our study was to investigate the role of skeletal muscle DNA methylation in combination with transcriptomic changes in obesity. Results Muscle biopsies were obtained basally from lean (n = 12; BMI = 23.4 ± 0.7 kg/m2) and obese (n = 10; BMI = 32.9 ± 0.7 kg/m2) participants in combination with euglycemic-hyperinsulinemic clamps to assess insulin sensitivity. We performed reduced representation bisulfite sequencing (RRBS) next-generation methylation and microarray analyses on DNA and RNA isolated from vastus lateralis muscle biopsies. There were 13,130 differentially methylated cytosines (DMC; uncorrected P < 0.05) that were altered in the promoter and untranslated (5' and 3'UTR) regions in the obese versus lean analysis. Microarray analysis revealed 99 probes that were significantly (corrected P < 0.05) altered. Of these, 12 genes (encompassing 22 methylation sites) demonstrated a negative relationship between gene expression and DNA methylation. Specifically, sorbin and SH3 domain containing 3 (SORBS3) which codes for the adapter protein vinexin was significantly decreased in gene expression (fold change −1.9) and had nine DMCs that were significantly increased in methylation in obesity (methylation differences ranged from 5.0 to 24.4 %). Moreover, differentially methylated region (DMR) analysis identified a region in the 5'UTR (Chr.8:22,423,530–22,423,569) of SORBS3 that was increased in methylation by 11.2 % in the obese group. The negative relationship observed between DNA methylation and gene expression for SORBS3 was validated by a site-specific sequencing approach, pyrosequencing, and qRT-PCR. Additionally, we performed transcription factor binding analysis and identified a number of transcription factors whose binding to the differentially methylated sites or region may contribute to obesity. Conclusions These results demonstrate that obesity alters the epigenome through DNA methylation and highlights novel transcriptomic changes in SORBS3 in skeletal muscle. Electronic supplementary material The online version of this article (doi:10.1186/s13148-016-0246-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Samantha E Day
- School of Life Sciences, Arizona State University, Tempe, AZ USA
| | - Richard L Coletta
- School for the Science of Health Care Delivery, Arizona State University, Phoenix, AZ USA
| | - Joon Young Kim
- Division of Weight Management and Wellness Children's Hospital of Pittsburgh, Pittsburgh, PA USA
| | | | - Tonya R Benjamin
- Endocrinology Department, Mayo Clinic in Arizona, Scottsdale, AZ USA
| | - Lori R Roust
- Endocrinology Department, Mayo Clinic in Arizona, Scottsdale, AZ USA
| | | | - Valentin Dinu
- The Department of Biomedical Informatics, Arizona State University, Phoenix, AZ USA
| | - Gabriel Q Shaibi
- College of Nursing and Health Innovation Arizona State University, Phoenix, AZ USA ; Mayo/ASU Center for Metabolic and Vascular Biology, Mayo Clinic in Arizona, Scottsdale, AZ USA
| | - Lawrence J Mandarino
- Division of Endocrinology, Diabetes and Metabolism in the Department of Medicine at the UA College of Medicine, University of Arizona, Tucson, AZ USA
| | - Dawn K Coletta
- Mayo/ASU Center for Metabolic and Vascular Biology, Mayo Clinic in Arizona, Scottsdale, AZ USA ; School of Nutrition and Health Promotion, College of Health Solutions, Arizona State University, 550 N. 3rd Street, Phoenix, AZ 85004 USA ; Department of Basic Medical Sciences, The University of Arizona College of Medicine, Phoenix, AZ USA
| |
Collapse
|
783
|
Reynard LN, Ratnayake M, Santibanez-Koref M, Loughlin J. Functional Characterization of the Osteoarthritis Susceptibility Mapping to CHST11-A Bioinformatics and Molecular Study. PLoS One 2016; 11:e0159024. [PMID: 27391021 PMCID: PMC4938163 DOI: 10.1371/journal.pone.0159024] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Accepted: 06/24/2016] [Indexed: 02/07/2023] Open
Abstract
The single nucleotide polymorphism (SNP) rs835487 is associated with hip osteoarthritis (OA) at the genome-wide significance level and is located within CHST11, which codes for carbohydrate sulfotransferase 11. This enzyme post-translationally modifies proteoglycan prior to its deposition in the cartilage extracellular matrix. Using bioinformatics and experimental analyses, our aims were to characterise the rs835487 association signal and to identify the causal functional variant/s. Database searches revealed that rs835487 resides within a linkage disequilibrium (LD) block of only 2.7 kb and is in LD (r2 ≥ 0.8) with six other SNPs. These are all located within intron 2 of CHST11, in a region that has predicted enhancer activity and which shows a high degree of conservation in primates. Luciferase reporter assays revealed that of the seven SNPs, rs835487 and rs835488, which have a pairwise r2 of 0.962, are the top functional candidates; the haplotype composed of the OA-risk conferring G allele of rs835487 and the corresponding T allele of rs835488 (the G-T haplotype) demonstrated significantly different enhancer activity relative to the haplotype composed of the non-risk A allele of rs835487 and the corresponding C allele of rs835488 (the A-C haplotype) (p < 0.001). Electrophoretic mobility shift assays and supershifts identified several transcription factors that bind more strongly to the risk-conferring G and T alleles of the two SNPs, including SP1, SP3, YY1 and SUB1. CHST11 was found to be upregulated in OA versus non-OA cartilage (p < 0.001) and was expressed dynamically during chondrogenesis. Its expression in adult cartilage did not however correlate with rs835487 genotype. Our data demonstrate that the OA susceptibility is mediated by differential protein binding to the alleles of rs835487 and rs835488, which are located within an enhancer whose target may be CHST11 during chondrogenesis or an alternative gene.
Collapse
Affiliation(s)
- Louise N. Reynard
- Musculoskeletal Research Group, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom
- * E-mail:
| | - Madhushika Ratnayake
- Musculoskeletal Research Group, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Mauro Santibanez-Koref
- Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - John Loughlin
- Musculoskeletal Research Group, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom
| |
Collapse
|
784
|
Zhang C, Xue P, Gao L, Chen X, Lin K, Yang X, Dai Y, Xu EY. Highly conserved epigenetic regulation of BOULE and DAZL is associated with human fertility. FASEB J 2016; 30:3424-3440. [PMID: 27358391 DOI: 10.1096/fj.201500167r] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Accepted: 06/21/2016] [Indexed: 11/11/2022]
Abstract
Separation of germ cells from somatic cells is a widespread feature of animal sexual reproduction, with a core set of germ cell factors conserved among diverse animals. It is not known what controls their conserved gonad-specific expression. Core components of epigenetic machinery are ancient, but its role in conserved tissue expression regulation remains unexplored. We found that promoters of the reproductive genes BOULE and DAZL exhibit differential DNA methylation, consistent with their gonad-specific expression in humans and mice. Low or little promoter methylation from the testicular tissue is attributed to spermatogenic cells of various stages in the testis. Such differential DNA methylation is present in the orthologous promoters not only of other mammalian species, but also of chickens and fish, supporting a highly conserved epigenetic mechanism. Furthermore, hypermethylation of DAZL and BOULE promoters in human sperm is associated with human infertility. Our data strongly suggest that epigenetic regulation may underlie conserved germ-cell-specific expression, and such a mechanism may play an important role in human fertility.-Zhang, C., Xue, P., Gao, L., Chen, X., Lin, K., Yang, X., Dai, Y., Xu, E. Y. Highly conserved epigenetic regulation of BOULE and DAZL is associated with human fertility.
Collapse
Affiliation(s)
- Chenwang Zhang
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China; and
| | - Peng Xue
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China; and Department of Urology, First Affiliated Hospital, Nanjing Medical University, Nanjing, China
| | - Liuze Gao
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China; and
| | - Xia Chen
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China; and
| | - Kaibo Lin
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China; and
| | - Xiaoyu Yang
- Department of Urology, First Affiliated Hospital, Nanjing Medical University, Nanjing, China
| | - Yifan Dai
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China; and
| | - Eugene Yujun Xu
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China; and
| |
Collapse
|
785
|
Ng TK, Liang XY, Lai TYY, Ma L, Tam POS, Wang JX, Chen LJ, Chen H, Pang CP. HTRA1 promoter variant differentiates polypoidal choroidal vasculopathy from exudative age-related macular degeneration. Sci Rep 2016; 6:28639. [PMID: 27338780 PMCID: PMC4919652 DOI: 10.1038/srep28639] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Accepted: 06/07/2016] [Indexed: 11/15/2022] Open
Abstract
Exudative age-related macular degeneration (AMD) and polypoidal choroidal vasculopathy (PCV) share similar abnormal choroidal vasculature, but responses to treatments are different. In this study, we sequenced the whole HTRA1 gene and its promoter by direct sequencing in a Hong Kong Chinese PCV cohort. We identified rs11200638, c.34delCinsTCCT, c.59C>T, rs1049331 and rs2293870 significantly associated with PCV. Notably, rs2672598 was significantly associated with exudative AMD (p = 1.31 × 10(-4)) than PCV (p = 0.11). Logistic regression indicated that rs2672598 (p = 2.27 × 10(-3)) remained significant after adjusting for rs11200638 in exudative AMD. Moreover, the rs11200638-rs2672598 joint genotype AA-CC conferred higher risk to exudative AMD (43.11 folds) than PCV (3.68 folds). Promoter analysis showed that rs2672598 C-allele showed higher luciferase expression than wildtype T-allele (p = 0.026), independent of rs11200638 genotype (p = 0.621). Coherently, vitreous humor HTRA1 expression with rs2672598 CC genotype was significantly higher than that with TT genotype by 2.56 folds (p = 0.02). Furthermore, rs2672598 C-allele was predicted to alter the transcription factor binding sites, but not rs11200638 A-allele. Our results revealed that HTRA1 rs2672598 is more significantly associated with exudative AMD than PCV in ARMS2/HTRA1 region, and it is responsible for elevated HTRA1 transcriptional activity and HTRA1 protein expression.
Collapse
Affiliation(s)
- Tsz Kin Ng
- Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Hong Kong
| | - Xiao Ying Liang
- Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Hong Kong
| | - Timothy Y. Y. Lai
- Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Hong Kong
| | - Li Ma
- Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Hong Kong
| | - Pancy O. S. Tam
- Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Hong Kong
| | - Jian Xiong Wang
- Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Hong Kong
| | - Li Jia Chen
- Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Hong Kong
| | - Haoyu Chen
- The Joint Shantou International Eye Center of Shantou University and The Chinese University of Hong Kong, Shantou, China
| | - Chi Pui Pang
- Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Hong Kong
| |
Collapse
|
786
|
HCK is a survival determinant transactivated by mutated MYD88, and a direct target of ibrutinib. Blood 2016; 127:3237-52. [DOI: 10.1182/blood-2016-01-695098] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2016] [Accepted: 04/20/2016] [Indexed: 12/15/2022] Open
Abstract
Key Points
HCK transcription and activation is triggered by mutated MYD88, and is an important determinant of pro-survival signaling. HCK is also a target of ibrutinib, and inhibition of its kinase activity triggers apoptosis in mutated MYD88 cells.
Collapse
|
787
|
Rao S, Lam MHB, Yeung VSY, Wing YK, Waye MMY. Association of HOMER1 rs2290639 with suicide attempts in Hong Kong Chinese and the potentially functional role of this polymorphism. SPRINGERPLUS 2016; 5:767. [PMID: 27386253 PMCID: PMC4912501 DOI: 10.1186/s40064-016-2404-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Accepted: 05/24/2016] [Indexed: 01/06/2023]
Abstract
OBJECTIVE Animal evidence and genetic studies suggest that HOMER1 (homer homolog 1) is involved in the etiology of suicidal behavior and major depression disorder (MDD). However, most of genetic studies were performed in Caucasians and the potentially functional role of associated polymorphisms in HOMER1 was seldom reported. The purpose of this study was to investigate the association of a HOMER1 polymorphism rs2290639 with suicide attempts (SA) and MDD in Hong Kong Chinese, and then briefly elucidate the potentially functional role of the associated polymorphism. METHODS NEO personality inventory, impulsiveness and depression rating scales were completed by the subjects. The association studies of HOMER1 rs2290639 with SA or MDD were performed by case-control association studies. The bioinformatics analyses were adapted to predict potential transcription factors binding sites for the associated polymorphism. RESULTS The association studies and meta-analysis suggested that the HOMER1 rs2290639 was significantly associated with susceptibility to SA but seemed not to be associated with MDD in Hong Kong Chinese. This polymorphism might affect the transcription of the HOMER1 gene through interacting with a reliable transcription factor as found by three of four bioinformatics tools. In addition, close correlations between impulsiveness and NEO personality five factors were found in SA and MDD patients, which provide a possible way to assess the impulsiveness of patients through subjects' personality profiles for Hong Kong Chinese. CONCLUSIONS The HOMER1 rs2290639 polymorphism was significantly associated with susceptibility to SA in Hong Kong Chinese affected by psychiatric disorders, which might be explained by the potentially functional role of this polymorphism.
Collapse
Affiliation(s)
- Shitao Rao
- />Croucher Laboratory for Human Genomics, Rm324A, Lo Kwee-Seong Integrated Biomedical Sciences Building, School of Biomedical Sciences, Area 39; The Nethersole School of Nursing, The Chinese University of Hong Kong, Shatin, N.T. Hong Kong
| | - Marco H. B. Lam
- />Department of Psychiatry, Shatin Hospital, The Chinese University of Hong Kong, 33 Ah Kong Kok Street, Shatin, N.T. Hong Kong
| | - Venus S. Y. Yeung
- />Croucher Laboratory for Human Genomics, Rm324A, Lo Kwee-Seong Integrated Biomedical Sciences Building, School of Biomedical Sciences, Area 39; The Nethersole School of Nursing, The Chinese University of Hong Kong, Shatin, N.T. Hong Kong
| | - Yun Kwok Wing
- />Department of Psychiatry, Shatin Hospital, The Chinese University of Hong Kong, 33 Ah Kong Kok Street, Shatin, N.T. Hong Kong
| | - Mary Miu Yee Waye
- />Croucher Laboratory for Human Genomics, Rm324A, Lo Kwee-Seong Integrated Biomedical Sciences Building, School of Biomedical Sciences, Area 39; The Nethersole School of Nursing, The Chinese University of Hong Kong, Shatin, N.T. Hong Kong
| |
Collapse
|
788
|
Allele-specific DNA methylation reinforces PEAR1 enhancer activity. Blood 2016; 128:1003-12. [PMID: 27313330 DOI: 10.1182/blood-2015-11-682153] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2015] [Accepted: 06/13/2016] [Indexed: 01/07/2023] Open
Abstract
Genetic variation in the PEAR1 locus is linked to platelet reactivity and cardiovascular disease. The major G allele of rs12041331, an intronic cytosine guanine dinucleotide-single-nucleotide polymorphism (CpG-SNP), is associated with higher PEAR1 expression in platelets and endothelial cells than the minor A allele. The molecular mechanism underlying this difference remains elusive. We have characterized the histone modification profiles of the intronic region surrounding rs12041331 and identified H3K4Me1 enhancer-specific enrichment for the region that covers the CpG-SNP. Interestingly, methylation studies revealed that the CpG site is fully methylated in leukocytes of GG carriers. Nuclear protein extracts from megakaryocytes, endothelial cells, vs control HEK-293 cells show a 3-fold higher affinity for the methylated G allele compared with nonmethylated G or A alleles in a gel electrophoretic mobility shift assay. To understand the positive relationship between methylation and gene expression, we studied DNA methylation at 4 different loci of PEAR1 during in vitro megakaryopoiesis. During differentiation, the CpG-SNP remained fully methylated, while we observed rapid methylation increases at the CpG-island overlapping the first 5'-untranslated region exon, paralleling the increased PEAR1 expression. In the same region, A-allele carriers of rs12041331 showed significantly lower DNA methylation at CGI1 compared with GG homozygote. This CpG-island contains binding sites for the methylation-sensitive transcription factor CTCF, whose binding is known to play a role in enhancer activation and/or repression. In conclusion, we report the molecular characterization of the first platelet function-related CpG-SNP, a genetic predisposition that reinforces PEAR1 enhancer activity through allele-specific DNA methylation.
Collapse
|
789
|
MGMT DNA repair gene promoter/enhancer haplotypes alter transcription factor binding and gene expression. Cell Oncol (Dordr) 2016; 39:435-447. [PMID: 27306526 DOI: 10.1007/s13402-016-0286-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/03/2016] [Indexed: 10/21/2022] Open
Abstract
BACKGROUND The O6-methylguanine-DNA methyltransferase (MGMT) protein removes O6-alkyl-guanine adducts from DNA. MGMT expression can thus alter the sensitivity of cells and tissues to environmental and chemotherapeutic alkylating agents. Previously, we defined the haplotype structure encompassing single nucleotide polymorphisms (SNPs) in the MGMT promoter/enhancer (P/E) region and found that haplotypes, rather than individual SNPs, alter MGMT promoter activity. The exact mechanism(s) by which these haplotypes exert their effect on MGMT promoter activity is currently unknown, but we noted that many of the SNPs comprising the MGMT P/E haplotypes are located within or in close proximity to putative transcription factor binding sites. Thus, these haplotypes could potentially affect transcription factor binding and, subsequently, alter MGMT promoter activity. METHODS In this study, we test the hypothesis that MGMT P/E haplotypes affect MGMT promoter activity by altering transcription factor (TF) binding to the P/E region. We used a promoter binding TF profiling array and a reporter assay to evaluate the effect of different P/E haplotypes on TF binding and MGMT expression, respectively. RESULTS Our data revealed a significant difference in TF binding profiles between the different haplotypes evaluated. We identified TFs that consistently showed significant haplotype-dependent binding alterations (p ≤ 0.01) and revealed their role in regulating MGMT expression using siRNAs and a dual-luciferase reporter assay system. CONCLUSIONS The data generated support our hypothesis that promoter haplotypes alter the binding of TFs to the MGMT P/E and, subsequently, affect their regulatory function on MGMT promoter activity and expression level.
Collapse
|
790
|
Lin AHY, Shang Y, Mitzner W, Sham JSK, Tang WY. Aberrant DNA Methylation of Phosphodiesterase [corrected] 4D Alters Airway Smooth Muscle Cell Phenotypes. Am J Respir Cell Mol Biol 2016; 54:241-9. [PMID: 26181301 DOI: 10.1165/rcmb.2015-0079oc] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Airway hyperresponsiveness (AHR) is a hallmark feature in asthma characterized by exaggerated airway contractile response to stimuli due to increased airway sensitivity and chronic airway remodeling. We have previously shown that allergen-induced AHR in mice is associated with aberrant DNA methylation in the lung genome, suggesting that AHR could be epigenetically regulated, and these changes might predispose the animals to asthma. Previous studies demonstrated that overexpression of phosphodiesterase 4D (PDE4D) is associated with increased AHR. However, epigenetic regulation of this gene in asthmatic airway smooth muscle cells (ASMCs) has not been examined. In this study, we aimed to examine the relationship between epigenetic regulation of PDE4D and ASMC phenotypes. We identified CpG site-specific hypomethylation at PDE4D promoter in human asthmatic ASMCs. We next used methylated oligonucleotides to introduce CpG site-specific methylation at PDE4D promoter and examined its effect on ASMCs. We showed that PDE4D methylation decreased cell proliferation and migration of asthmatic ASMCs. We further elucidated that methylated PDE4D decreased PDE4D expression in asthmatic ASMCs, increased cAMP level, and inhibited the aberrant increase in Ca(2+) level. Moreover, PDE4D methylation reduced the phosphorylation level of downstream effectors of Ca(2+) signaling, including myosin light chain kinase and p38. Taken together, our findings demonstrate that gene-specific epigenetic changes may predispose ASMCs to asthma through alterations in cell phenotypes. Modulation of ASMC phenotypes by methylated PDE4D oligonucleotides can reverse the aberrant ASMC functions to normal phenotypes. This has provided new insight to the development of novel therapeutic options for this debilitative disease.
Collapse
Affiliation(s)
- Amanda H Y Lin
- 1 Division of Pulmonary and Critical Care Medicine, Johns Hopkins University, Baltimore, Maryland; and
| | - Yan Shang
- 2 Department of Environmental Health Sciences, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Wayne Mitzner
- 2 Department of Environmental Health Sciences, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - James S K Sham
- 1 Division of Pulmonary and Critical Care Medicine, Johns Hopkins University, Baltimore, Maryland; and.,2 Department of Environmental Health Sciences, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Wan-yee Tang
- 2 Department of Environmental Health Sciences, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| |
Collapse
|
791
|
NF-κB-Regulated miR-99a Modulates Endothelial Cell Inflammation. Mediators Inflamm 2016; 2016:5308170. [PMID: 27403035 PMCID: PMC4923609 DOI: 10.1155/2016/5308170] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Accepted: 05/15/2016] [Indexed: 11/18/2022] Open
Abstract
Objective. The present study was performed to investigate the effects and mechanisms of miR-99a on LPS-induced endothelial cell inflammation, as well as the regulation of NF-κB on miR-99a production. Methods and Results. ELISA showed that LPS treatment significantly promoted the secretion of inflammatory factors (TNF-α, IL-6, IL-1β, and MCP-1). LPS treatment also inhibited miR-99a production and promoted mTOR expression and NF-κB nuclear translocation. Overexpression of miR-99a suppressed the LPS-induced TNF-α, IL-6, IL-1β, and MCP-1 overproduction, mTOR upregulation, and NF-κB nuclear translocation. The PROMO software analysis indicated NF-κB binding site in the −1643 to −1652 region of miR-99a promoter. Dual luciferase reporter analysis, electrophoretic mobility shift assays (EMSA), and chromosome immunoprecipitation (ChIP) assays demonstrated that NF-κB promoted the transcription of miR-99a by binding to the −1643 to −1652 region of miR-99a promoter. Further studies on HUVECs verified the regulatory effects of NF-κB on miR-99a production. Conclusion. MiR-99a inhibited the LPS-induced HUVECs inflammation via inhibition of the mTOR/NF-κB signal. NF-κB promoted miR-99a production by binding to the −1643 to −1652 region of miR-99a promoter. Considering the importance of endothelial inflammation on cardiovascular diseases, such as atherosclerosis, our results may provide a new insight into the pathogenesis and therapy of atherosclerosis.
Collapse
|
792
|
Fekry B, Jeffries KA, Esmaeilniakooshkghazi A, Ogretmen B, Krupenko SA, Krupenko NI. CerS6 Is a Novel Transcriptional Target of p53 Protein Activated by Non-genotoxic Stress. J Biol Chem 2016; 291:16586-96. [PMID: 27302066 PMCID: PMC4974374 DOI: 10.1074/jbc.m116.716902] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Indexed: 12/19/2022] Open
Abstract
Our previous study suggested that ceramide synthase 6 (CerS6), an enzyme in sphingolipid biosynthesis, is regulated by p53: CerS6 was elevated in several cell lines in response to transient expression of p53 or in response to folate stress, which is known to activate p53. It was not clear, however, whether CerS6 gene is a direct transcriptional target of p53 or whether this was an indirect effect through additional regulatory factors. In the present study, we have shown that the CerS6 promoter is activated by p53 in luciferase assays, whereas transcriptionally inactive R175H p53 mutant failed to induce the luciferase expression from this promoter. In vitro immunoprecipitation assays and gel shift analyses have further demonstrated that purified p53 binds within the CerS6 promoter sequence spanning 91 bp upstream and 60 bp downstream of the transcription start site. The Promo 3.0.2 online tool for the prediction of transcription factor binding sites indicated the presence of numerous putative non-canonical p53 binding motifs in the CerS6 promoter. Luciferase assays and gel shift analysis have identified a single motif upstream of the transcription start as a key p53 response element. Treatment of cells with Nutlin-3 or low concentrations of actinomycin D resulted in a strong elevation of CerS6 mRNA and protein, thus demonstrating that CerS6 is a component of the non-genotoxic p53-dependent cellular stress response. This study has shown that by direct transcriptional activation of CerS6, p53 can regulate specific ceramide biosynthesis, which contributes to the pro-apoptotic cellular response.
Collapse
Affiliation(s)
- Baharan Fekry
- From the Nutrition Research Institute, University of North Carolina at Chapel Hill, Kannapolis, North Carolina 28081
| | - Kristen A Jeffries
- From the Nutrition Research Institute, University of North Carolina at Chapel Hill, Kannapolis, North Carolina 28081
| | - Amin Esmaeilniakooshkghazi
- From the Nutrition Research Institute, University of North Carolina at Chapel Hill, Kannapolis, North Carolina 28081
| | - Besim Ogretmen
- the Department of Biochemistry and Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina 29425, and
| | - Sergey A Krupenko
- From the Nutrition Research Institute, University of North Carolina at Chapel Hill, Kannapolis, North Carolina 28081, the Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599
| | - Natalia I Krupenko
- From the Nutrition Research Institute, University of North Carolina at Chapel Hill, Kannapolis, North Carolina 28081, the Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599
| |
Collapse
|
793
|
Bhakuni T, Sharma A, Ali MF, Mahapatra M, Saxena R, Jairajpuri MA. Identification of 2 Novel Polymorphisms and rs3138521 in 5′ Untranslated Region of SERPINC1 Gene in North Indian Population With Deep Vein Thrombosis. Clin Appl Thromb Hemost 2016; 23:865-870. [DOI: 10.1177/1076029616652725] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Antithrombin III (AT) is the most important endogenous anticoagulant, and genetic variability in SERPINC1, gene encoding AT, is low. Mutations leading to AT deficiency and increased thrombotic risk are well known; however, only 2 studies have reported mutations in regulatory region of SERPINC1 gene till date. Aim of the present study was to identify genetic variations in SERPINC1 5′ untranslated region (UTR) in Indian patients with deep vein thrombosis (DVT) having AT deficiency. DNA sequencing was used to identify underlying genetic defects in SERPINC1 regulatory region. In silico tools TFBIND and PROMO were used to identify transcription factor binding sites in the promoter region. We have identified 2 novel polymorphisms, g.25G>A and g.−1A>T, and 2 known g.67G>A and rs3138521 5′ UTR polymorphisms in SERPINC1 regulatory region in Indian patients with DVT for the first time. In present study, allele frequencies of rs3138521 (S: 0.37 and F: 0.63) were similar to that reported in Western population and were not associated with low plasma AT levels ( P value .5). This is the first report of regulatory region polymorphisms in SERPINC1 gene in Indian population. Our results strongly suggest that similar studies should be included when ever no mutation is detected in protein-coding region of AT gene.
Collapse
Affiliation(s)
- Teena Bhakuni
- Protein Conformation and Enzymology Lab, Department of Biosciences, Jamia Millia Islamia, New Delhi, India
| | - Amit Sharma
- Department of Hematology, All India Institute of Medical Sciences, New Delhi, India
| | - Mohammad Farhan Ali
- Protein Conformation and Enzymology Lab, Department of Biosciences, Jamia Millia Islamia, New Delhi, India
| | - Manoranjan Mahapatra
- Department of Hematology, All India Institute of Medical Sciences, New Delhi, India
| | - Renu Saxena
- Department of Hematology, All India Institute of Medical Sciences, New Delhi, India
| | - Mohamad Aman Jairajpuri
- Protein Conformation and Enzymology Lab, Department of Biosciences, Jamia Millia Islamia, New Delhi, India
| |
Collapse
|
794
|
Mesic A, Rogar M, Hudler P, Juvan R, Komel R. Association of the AURKA and AURKC gene polymorphisms with an increased risk of gastric cancer. IUBMB Life 2016; 68:634-44. [PMID: 27270838 DOI: 10.1002/iub.1521] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Accepted: 05/18/2016] [Indexed: 12/19/2022]
Abstract
Single nucleotide polymorphisms (SNPs) in mitotic checkpoint genes can contribute to susceptibility of human cancer, including gastric cancer (GC). We aimed to investigate the effects of Aurora kinase A (AURKA), Aurora kinase B (AURKB), and Aurora kinase C (AURKC) gene polymorphisms on GC risk in Slovenian population. We genotyped four SNPs in AURKA (rs2273535 and rs1047972), AURKB (rs2241909), and AURKC (rs758099) in a total of 128 GC patients and 372 healthy controls using TaqMan allelic discrimination assays to evaluate their effects on GC risk. Our results showed that genotype frequencies between cases and controls were significantly different for rs1047972 and rs758099 (P < 0.05). Our study demonstrated that AURKA rs1047972 TT and (CC + CT) genotypes were significantly associated with an increased risk of gastric cancer. Our results additionally revealed that AURKC rs758099 TT and (CC + CT) genotypes were also associated with increased GC risk. In stratified analysis, genotypes TT and (CC + CT) of AURKA rs1047972 SNP were associated with increased risk of both, intestinal and diffuse, types of GC. In addition, AURKC rs758099 TT and (CC + CT) genotypes were positively associated with increased intestinal type GC risk, but not with an increased diffuse type GC risk. Based on these results, we can conclude that AURKA rs1047972 and AURKC rs758099 polymorphisms could affect the risk of GC development. Further larger studies are needed to confirm these findings. © 2016 IUBMB Life, 68(8):634-644, 2016.
Collapse
Affiliation(s)
- Aner Mesic
- Department of Biology, Faculty of Science, University of Sarajevo, Sarajevo, Bosnia and Herzegovina
| | - Marija Rogar
- Medical Centre for Molecular Biology, Institute of Biochemistry, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Petra Hudler
- Medical Centre for Molecular Biology, Institute of Biochemistry, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Robert Juvan
- Clinical Department for Abdominal Surgery, University Medical Centre Ljubljana, Ljubljana, Slovenia
| | - Radovan Komel
- Medical Centre for Molecular Biology, Institute of Biochemistry, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| |
Collapse
|
795
|
Suzuki T, Swift LL. Discovery of Novel Splice Variants and Regulatory Mechanisms for Microsomal Triglyceride Transfer Protein in Human Tissues. Sci Rep 2016; 6:27308. [PMID: 27256115 PMCID: PMC4891672 DOI: 10.1038/srep27308] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Accepted: 05/16/2016] [Indexed: 11/16/2022] Open
Abstract
Microsomal triglyceride transfer protein (MTP) is a unique lipid transfer protein essential for the assembly of triglyceride-rich lipoproteins by the liver and intestine. Previous studies in mice identified a splice variant of MTP with an alternate first exon. Splice variants of human MTP have not been reported. Using PCR approaches we have identified two splice variants in human tissues, which we have named MTP-B and MTP-C. MTP-B has a unique first exon (Ex1B) located 10.5 kb upstream of the first exon (Ex1A) for canonical MTP (MTP-A); MTP-C contains both first exons for MTP-A and MTP-B. MTP-B was found in a number of tissues, whereas MTP-C was prominent in brain and testis. MTP-B does not encode a protein; MTP-C encodes the same protein encoded by MTP-A, although MTP-C translation is strongly inhibited by regulatory elements within its 5′-UTR. Using luciferase assays, we demonstrate that the promoter region upstream of exon 1B is quite adequate to drive expression of MTP. We conclude that alternate splicing plays a key role in regulating cellular MTP levels by introducing distinct promoter regions and unique 5′-UTRs, which contain elements that alter translation efficiency, enabling the cell to optimize MTP activity.
Collapse
Affiliation(s)
- Takashi Suzuki
- Department of Pathology, Microbiology and Immunology Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Larry L Swift
- Department of Pathology, Microbiology and Immunology Vanderbilt University School of Medicine, Nashville, TN 37232, USA.,Research Service, Veterans Affairs, Tennessee Valley Health Care System, Nashville, TN, USA
| |
Collapse
|
796
|
Samblas M, Milagro FI, Gómez-Abellán P, Martínez JA, Garaulet M. Methylation on the Circadian Gene BMAL1 Is Associated with the Effects of a Weight Loss Intervention on Serum Lipid Levels. J Biol Rhythms 2016; 31:308-17. [PMID: 26873744 DOI: 10.1177/0748730416629247] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The circadian clock system has been linked to the onset and development of obesity and some accompanying comorbidities. Epigenetic mechanisms, such as DNA methylation, are putatively involved in the regulation of the circadian clock system. The aim of this study was to investigate the influence of a weight loss intervention based on an energy-controlled Mediterranean dietary pattern in the methylation levels of 3 clock genes, BMAL1, CLOCK, and NR1D1, and the association between the methylation levels and changes induced in the serum lipid profile with the weight loss treatment. The study sample enrolled 61 women (body mass index = 28.6 ± 3.4 kg/m(2); age: 42.2 ± 11.4 years), who followed a nutritional program based on a Mediterranean dietary pattern. DNA was isolated from whole blood obtained at the beginning and end point. Methylation levels at different CpG sites of BMAL1, CLOCK, and NR1D1 were analyzed by Sequenom's MassArray. The energy-restricted intervention modified the methylation levels of different CpG sites in BMAL1 (CpGs 5, 6, 7, 9, 11, and 18) and NR1D1 (CpGs 1, 10, 17, 18, 19, and 22). Changes in cytosine methylation in the CpG 5 to 9 region of BMAL1 with the intervention positively correlated with the eveningness profile (p = 0.019). The baseline methylation of the CpG 5 to 9 region in BMAL1 positively correlated with energy (p = 0.047) and carbohydrate (p = 0.017) intake and negatively correlated with the effect of the weight loss intervention on total cholesterol (p = 0.032) and low-density lipoprotein cholesterol (p = 0.005). Similar significant and positive correlations were found between changes in methylation levels in the CpG 5 to 9 region of BMAL1 due to the intervention and changes in serum lipids (p < 0.05). This research describes apparently for the first time an association between changes in the methylation of the BMAL1 gene with the intervention and the effects of a weight loss intervention on blood lipids levels.
Collapse
Affiliation(s)
- Mirian Samblas
- Department of Nutrition, Food Science and Physiology, University of Navarra, Pamplona, Spain
| | - Fermin I Milagro
- Department of Nutrition, Food Science and Physiology, University of Navarra, Pamplona, Spain Centre for Nutrition Research, University of Navarra, Pamplona, Spain CIBERobn, Physiopathology of Obesity, Carlos III Institute, Madrid, Spain
| | | | - J Alfredo Martínez
- Department of Nutrition, Food Science and Physiology, University of Navarra, Pamplona, Spain Centre for Nutrition Research, University of Navarra, Pamplona, Spain CIBERobn, Physiopathology of Obesity, Carlos III Institute, Madrid, Spain IdiSNA, Navarra's Health Research Institute, Pamplona, Spain
| | - Marta Garaulet
- Department of Physiology, Faculty of Biology, University of Murcia, IMIB, Murcia, Spain
| |
Collapse
|
797
|
Farkas AE, Hilgarth RS, Capaldo CT, Gerner-Smidt C, Powell DR, Vertino PM, Koval M, Parkos CA, Nusrat A. HNF4α regulates claudin-7 protein expression during intestinal epithelial differentiation. THE AMERICAN JOURNAL OF PATHOLOGY 2016. [PMID: 26216285 DOI: 10.1016/j.ajpath.2015.04.023] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
The intestinal epithelium is a dynamic barrier that maintains the distinct environments of intestinal tissue and lumen. Epithelial barrier function is defined principally by tight junctions, which, in turn, depend on the regulated expression of claudin family proteins. Claudins are expressed differentially during intestinal epithelial cell (IEC) differentiation. However, regulatory mechanisms governing claudin expression during epithelial differentiation are incompletely understood. We investigated the molecular mechanisms regulating claudin-7 during IEC differentiation. Claudin-7 expression is increased as epithelial cells differentiate along the intestinal crypt-luminal axis. By using model IECs we observed increased claudin-7 mRNA and nascent heteronuclear RNA levels during differentiation. A screen for potential regulators of the CLDN7 gene during IEC differentiation was performed using a transcription factor/DNA binding array, CLDN7 luciferase reporters, and in silico promoter analysis. We identified hepatocyte nuclear factor 4α as a regulatory factor that bound endogenous CLDN7 promoter in differentiating IECs and stimulated CLDN7 promoter activity. These findings support a role of hepatocyte nuclear factor 4α in controlling claudin-7 expression during IEC differentiation.
Collapse
Affiliation(s)
- Attila E Farkas
- Epithelial Pathobiology and Mucosal Inflammation Research Unit, Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Roland S Hilgarth
- Epithelial Pathobiology and Mucosal Inflammation Research Unit, Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Christopher T Capaldo
- Epithelial Pathobiology and Mucosal Inflammation Research Unit, Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Christian Gerner-Smidt
- Epithelial Pathobiology and Mucosal Inflammation Research Unit, Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Doris R Powell
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Cell Biology, Emory University School of Medicine, Atlanta, Georgia
| | - Paula M Vertino
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Cell Biology, Emory University School of Medicine, Atlanta, Georgia
| | - Michael Koval
- Department of Radiation Oncology and the Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia
| | - Charles A Parkos
- Epithelial Pathobiology and Mucosal Inflammation Research Unit, Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia; Department of Pathology, University of Michigan, Ann Arbor, Michigan
| | - Asma Nusrat
- Epithelial Pathobiology and Mucosal Inflammation Research Unit, Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia; Department of Pathology, University of Michigan, Ann Arbor, Michigan.
| |
Collapse
|
798
|
Huang TT, Ping YH, Wang AM, Ke CC, Fang WL, Huang KH, Lee HC, Chi CW, Yeh TS. The reciprocal regulation loop of Notch2 pathway and miR-23b in controlling gastric carcinogenesis. Oncotarget 2016; 6:18012-26. [PMID: 26041881 PMCID: PMC4627232 DOI: 10.18632/oncotarget.4000] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Accepted: 05/06/2015] [Indexed: 01/03/2023] Open
Abstract
Gastric carcinoma is one of the most common malignancies and the third highest cause of global cancer-related death. Notch2 receptor intracellular domain (N2IC), the activated form of Notch2 receptor, enhances gastric carcinogenesis. MicroRNAs (miRNAs) act as either oncogenes or tumor suppressors in tumorigenesis and cross-talk with Notch pathways. Herein, microRNA-23b (miR-23b) was identified as a Notch2 receptor-related miRNA and its role in gastric carcinogenesis was investigated. Levels of miR-23b in stomach adenocarcinoma samples were down-regulated, whereas those of Notch2 receptor, v-ets erythroblastosis virus E26 oncogene homolog 1 (Ets1), and E2F1 transcripts were up-regulated. Results also showed that N2IC down-regulated miR-23b expression in gastric cancer cells through up-regulating E2F1. The miR-23b inhibited gastric tumorigenesis including growth, viability, epithelial-mesenchymal transition, and abilities of colony formation, migration, invasion, and tumorsphere formation. Mechanistically, miR-23b suppressed tumor progression and pluripotency gene expression and affected tumorsphere ultra-structure in gastric cancer cells via targeting Notch2 receptor or Ets1. Furthermore, miR-23b diminished the xenografted tumor growth and lung metastasis of SC-M1 gastric cancer cells through Notch2 pathway. Our results suggest that Notch2 pathway and miR-23b interplay in a reciprocal regulation loop in gastric cancer cells and this axis plays an important role in gastric carcinogenesis.
Collapse
Affiliation(s)
- Tzu-Ting Huang
- Department and Institute of Pharmacology, School of Medicine, National Yang-Ming University, Taipei, Taiwan.,Department of Anatomy and Cell Biology, School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Yueh-Hsin Ping
- Department and Institute of Pharmacology, School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - An-Ming Wang
- Department of Anatomy and Cell Biology, School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Chia-Chi Ke
- Department of Anatomy and Cell Biology, School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Wen-Liang Fang
- Institute of Clinical Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan.,Department of Surgery, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Kuo-Hung Huang
- Institute of Clinical Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan.,Department of Surgery, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Hsin-Chen Lee
- Department and Institute of Pharmacology, School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Chin-Wen Chi
- Department and Institute of Pharmacology, School of Medicine, National Yang-Ming University, Taipei, Taiwan.,Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Tien-Shun Yeh
- Department of Anatomy and Cell Biology, School of Medicine, National Yang-Ming University, Taipei, Taiwan.,Institute of Biochemistry and Molecular Biology, National Yang-Ming University, Taipei, Taiwan.,Genome Research Center, National Yang-Ming University, Taipei, Taiwan.,Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
799
|
Falkenberg N, Anastasov N, Schaub A, Radulovic V, Schmitt M, Magdolen V, Aubele M. Secreted uPAR isoform 2 (uPAR7b) is a novel direct target of miR-221. Oncotarget 2016; 6:8103-14. [PMID: 25797271 PMCID: PMC4480738 DOI: 10.18632/oncotarget.3516] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2014] [Accepted: 02/03/2015] [Indexed: 02/01/2023] Open
Abstract
miR-221/-222 and components of the urokinase-type plasminogen activator system (uPAS) are associated with metastasis and poor prognosis in breast cancer, including the triple-negative subtype (TNBC). Modification of components of uPAS and involved miRNAs may contribute to targeted therapy for breast cancer patients. miR-221−/−222-overexpressing or miR-221-depleted cells were employed for qRT-PCR and Western blots to show associations of uPAR with miR-221/-222. To substantiate direct targeting of miR-221/-222 within 3′ UTR of the uPAR isoform 2, in silico analysesand in vitro assays were conducted. Significant associations between miR-221 and uPAR isoform 2 expressions were observed at the mRNA and protein levels in breast cancer cells representing TNBC. For the first time, the uPAR isoform 2 was demonstrated as direct target for miR-221/-222. Inhibition of miR-221 reduced uPAR protein expression and expression of the tumor cell invasion markers vimentin and RHOC. These results demonstrate a direct and positive regulation of the secreted uPAR isoform 2 by miR-221, increasing its protein expression, a prerequisite for malignancy, while the other uPAR isoforms (1, 3 and 4) are indirectly regulated through miR-10b and miR-221/-222. By targeting uPAR isoforms and/or miRNA-221/-222, the diagnosis and therapy of breast cancer, in particular in TNBC, could be significantly improved.
Collapse
Affiliation(s)
- Natalie Falkenberg
- Institute of Pathology, German Research Center for Environmental Health, Neuherberg, Germany
| | - Nataša Anastasov
- Institute of Radiation Biology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Annalisa Schaub
- Institute of Pathology, German Research Center for Environmental Health, Neuherberg, Germany
| | - Vanja Radulovic
- Institute of Radiation Biology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Manfred Schmitt
- Clinical Research Unit, Department of Obstetrics and Gynecology, Technische Universität München, München, Germany
| | - Viktor Magdolen
- Clinical Research Unit, Department of Obstetrics and Gynecology, Technische Universität München, München, Germany
| | - Michaela Aubele
- Institute of Pathology, German Research Center for Environmental Health, Neuherberg, Germany
| |
Collapse
|
800
|
Kannambath S. Micro-RNA Feedback Loops Modulating the Calcineurin/NFAT Signaling Pathway. Noncoding RNA 2016; 2:E3. [PMID: 29657261 PMCID: PMC5831902 DOI: 10.3390/ncrna2020003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Revised: 04/22/2016] [Accepted: 05/09/2016] [Indexed: 02/07/2023] Open
Abstract
Nuclear factor of activated T cells (NFAT) is a family of transcription factors important for innate and adaptive immune responses. NFAT activation is tightly regulated through the calcineurin/NFAT signaling pathway. There is increasing evidence on non-coding RNAs such as miRNAs playing a crucial role in regulating transcription factors and signaling pathways. However, not much is known about microRNAs (miRNAs) targeting the calcineurin/NFAT signaling pathway involved in immune response in human. In this study, a comprehensive pathway level analysis has been carried out to identify miRNAs regulating the calcineurin/NFAT signaling pathway. Firstly, by incorporating experimental data and computational predictions, 191 unique miRNAs were identified to be targeting the calcineurin/NFAT signaling pathway in humans. Secondly, combining miRNA expression data from activated T cells and computational predictions, 32 miRNAs were observed to be induced by NFAT transcription factors. Finally, 11 miRNAs were identified to be involved in a feedback loop to modulate the calcineurin/NFAT signaling pathway activity. This data demonstrate the potential role of miRNAs as regulators of the calcineurin/NFAT signaling pathway. The present study thus emphasizes the importance of pathway level analysis to identify miRNAs and understands their role in modulating signaling pathways and transcription factor activity.
Collapse
Affiliation(s)
- Shichina Kannambath
- Infection and Immunity, St. George's University of London, Cranmer Terrace, London SW17 0RE, UK.
| |
Collapse
|