1
|
Li S, Meng W, Guo Z, Liu M, He Y, Li Y, Ma Z. The miR-183 Cluster: Biogenesis, Functions, and Cell Communication via Exosomes in Cancer. Cells 2023; 12:1315. [PMID: 37174715 PMCID: PMC10177187 DOI: 10.3390/cells12091315] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 04/18/2023] [Accepted: 04/25/2023] [Indexed: 05/15/2023] Open
Abstract
Cancer is one of the leading causes of human death. MicroRNAs have been found to be closely associated with cancer. The miR-183 cluster, comprising miR-183, miR-96, and miR-182, is transcribed as a polycistronic miRNA cluster. Importantly, in most cases, these clusters promote cancer development through different pathways. Exosomes, as extracellular vesicles, play an important role in cellular communication and the regulation of the tissue microenvironment. Interestingly, the miR-183 cluster can be detected in exosomes and plays a functional regulatory role in tumor development. Here, the biogenesis and functions of the miR-183 cluster in highly prevalent cancers and their relationship with other non-coding RNAs are summarized. In addition, the miR-183 cluster in exosomes has also been discussed. Finally, we discuss the miR-183 cluster as a promising target for cancer therapy. This review is expected to provide a new direction for cancer treatment.
Collapse
Affiliation(s)
- Shuhui Li
- Lab for Noncoding RNA & Cancer, School of Life Sciences, Shanghai University, Shanghai 200444, China
| | - Wei Meng
- Lab for Noncoding RNA & Cancer, School of Life Sciences, Shanghai University, Shanghai 200444, China
| | - Ziyi Guo
- Lab for Noncoding RNA & Cancer, School of Life Sciences, Shanghai University, Shanghai 200444, China
| | - Min Liu
- Lab for Noncoding RNA & Cancer, School of Life Sciences, Shanghai University, Shanghai 200444, China
| | - Yanyun He
- Experimental Center of Life Sciences, Shanghai University, Shanghai 200444, China
| | - Yanli Li
- Lab for Noncoding RNA & Cancer, School of Life Sciences, Shanghai University, Shanghai 200444, China
| | - Zhongliang Ma
- Lab for Noncoding RNA & Cancer, School of Life Sciences, Shanghai University, Shanghai 200444, China
| |
Collapse
|
2
|
Keller DM, Perez IG. Dual regulation of miR-375 and CREM genes in pancreatic beta cells. Islets 2022; 14:139-148. [PMID: 35377267 PMCID: PMC8986308 DOI: 10.1080/19382014.2022.2060688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
MicroRNA-375 (miR-375) is upregulated in the islets of some diabetics and is correlated with poor outcome. Previous work in our laboratory showed that cyclic adenosine monophosphate (cAMP) reduces miR-375 expression and could provide a way to restore normal miR-375 levels, however the transcription repression mechanism is unknown. Using a chromatin immunoprecipitation assay we show that cAMP response element modulator (CREM) binds to the miR-375 promoter 3-fold above background and we find that CREM represses transcription from the miR-375 promoter 1.8-fold. While investigating miR-375 target genes we discovered that several microRNA:mRNA target prediction algorithms listed human CREM as a target gene of miR-375. The predicted binding site is conserved in primates but not in other species. We found that indeed miR-375 binds to the predicted site on human CREM and represses translation of a green fluorescent protein reporter gene by 30%. These findings suggest a primate-specific double-negative feedback loop, a mechanism that would keep these important β-cell regulators in check.
Collapse
Affiliation(s)
- David M. Keller
- Department of Biological Sciences, California State University Chico, Chico, CA, USA
- CONTACT David M. Keller Department of Biological Sciences, California State University, Chico, 900 W. 1st St, Chico, CA95929 linkedin.com/in/keller-david-6529485b
| | - Isis G. Perez
- Department of Biological Sciences, California State University Chico, Chico, CA, USA
| |
Collapse
|
3
|
Multi-Omics Analysis of Key microRNA-mRNA Metabolic Regulatory Networks in Skeletal Muscle of Obese Rabbits. Int J Mol Sci 2021; 22:ijms22084204. [PMID: 33921578 PMCID: PMC8072691 DOI: 10.3390/ijms22084204] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 04/10/2021] [Accepted: 04/15/2021] [Indexed: 12/25/2022] Open
Abstract
microRNAs (miRNAs), small non-coding RNA with a length of about 22 nucleotides, are involved in the energy metabolism of skeletal muscle cells. However, their molecular mechanism of metabolism in rabbit skeletal muscle is still unclear. In this study, 16 rabbits, 8 in the control group (CON-G) and 8 in the experimental group (HFD-G), were chosen to construct an obese model induced by a high-fat diet fed from 35 to 70 days of age. Subsequently, 54 differentially expressed miRNAs, 248 differentially expressed mRNAs, and 108 differentially expressed proteins related to the metabolism of skeletal muscle were detected and analyzed with three sequencing techniques (small RNA sequencing, transcriptome sequencing, and tandem mass tab (TMT) protein technology). It was found that 12 miRNAs and 12 core genes (e.g., CRYL1, VDAC3 and APIP) were significantly different in skeletal muscle from rabbits in the two groups. The network analysis showed that seven miRNA-mRNA pairs were involved in metabolism. Importantly, two miRNAs (miR-92a-3p and miR-30a/c/d-5p) regulated three transcription factors (MYBL2, STAT1 and IKZF1) that may be essential for lipid metabolism. These results enhance our understanding of molecular mechanisms associated with rabbit skeletal muscle metabolism and provide a basis for future studies in the metabolic diseases of human obesity.
Collapse
|
4
|
Gerasymchuk D, Hubiernatorova A, Domanskyi A. MicroRNAs Regulating Cytoskeleton Dynamics, Endocytosis, and Cell Motility-A Link Between Neurodegeneration and Cancer? Front Neurol 2020; 11:549006. [PMID: 33240194 PMCID: PMC7680873 DOI: 10.3389/fneur.2020.549006] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Accepted: 10/06/2020] [Indexed: 12/13/2022] Open
Abstract
The cytoskeleton is one of the most mobile and complex cell structures. It is involved in cellular transport, cell division, cell shape formation and adaptation in response to extra- and intracellular stimuli, endo- and exocytosis, migration, and invasion. These processes are crucial for normal cellular physiology and are affected in several pathological processes, including neurodegenerative diseases, and cancer. Some proteins, participating in clathrin-mediated endocytosis (CME), play an important role in actin cytoskeleton reorganization, and formation of invadopodia in cancer cells and are also deregulated in neurodegenerative disorders. However, there is still limited information about the factors contributing to the regulation of their expression. MicroRNAs are potent negative regulators of gene expression mediating crosstalk between different cellular pathways in cellular homeostasis and stress responses. These molecules regulate numerous genes involved in neuronal differentiation, plasticity, and degeneration. Growing evidence suggests the role of microRNAs in the regulation of endocytosis, cell motility, and invasiveness. By modulating the levels of such microRNAs, it may be possible to interfere with CME or other processes to normalize their function. In malignancy, the role of microRNAs is undoubtful, and therefore changing their levels can attenuate the carcinogenic process. Here we review the current advances in our understanding of microRNAs regulating actin cytoskeleton dynamics, CME and cell motility with a special focus on neurodegenerative diseases, and cancer. We investigate whether current literature provides an evidence that microRNA-mediated regulation of essential cellular processes, such as CME and cell motility, is conserved in neurons, and cancer cells. We argue that more research effort should be addressed to study the neuron-specific functions on microRNAs. Disease-associated microRNAs affecting essential cellular processes deserve special attention both from the view of fundamental science and as future neurorestorative or anti-cancer therapies.
Collapse
Affiliation(s)
- Dmytro Gerasymchuk
- Institute of Biotechnology, Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland
- Institute of Molecular Biology and Genetics, National Academy of Sciences of Ukraine, Kyiv, Ukraine
| | | | - Andrii Domanskyi
- Institute of Biotechnology, Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland
| |
Collapse
|
5
|
Exosomal miR-183-5p promotes angiogenesis in colorectal cancer by regulation of FOXO1. Aging (Albany NY) 2020; 12:8352-8371. [PMID: 32364530 PMCID: PMC7244076 DOI: 10.18632/aging.103145] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Accepted: 03/31/2020] [Indexed: 12/14/2022]
Abstract
Exosomes play important roles in proliferation and microenvironment modulation of many types of cancers, including colorectal cancer (CRC). However, the inhibitory effect of CRC cells-derived exosomes in angiogenesis has not been fully discussed. In this study, the roles of microRNA-183-5p (miR-183-5p) in abundant in exosomes secreted from the CRC cells were investigated. Initially, microarray analysis was employed to determine the differentially expressed miRNAs. Exosomes isolated from CRC cells were co-cultured with HMEC-1 cells to explore the role of exosomes in angiogenesis. Further, the effects of CRC cell-derived exosomal miR-183-5p on proliferation, invasion and tube formation abilities of HMEC-1 cells were assessed. The preventative effect of exosomal miR-183-5p in vivo was measured in nude mice. Initially, it was found that FOXO1 was downregulated while miR-183-5p was upregulated in CRC. Additionally, the inhibition of miR-183-5p was suggested to suppress proliferation, invasion and tube formation abilities of HMEC-1 cells through upregulating FOXO1. Then, in vitro assays demonstrated that CRC cell-derived exosomes overexpressing miR-183-5p contributed to an enhanced proliferation, invasion and tube formation abilities of HMEC-1 cells. Furthermore, in vivo experiments confirmed the tumor-promotive effects of CRC cell-derived exosomal miR-183-5p. Collectively, our study demonstrates that the CRC cell-derived exosomes overexpressing miR-183-5p aggravates CRC through the regulation of FOXO1. Exosomes overexpressing miR-183-5p might be a potential treatment biomarker for CRC.
Collapse
|
6
|
Lan X, Wu N, Wu L, Qu K, Osoro EK, Guan D, Du X, Wang B, Chen S, Miao J, Ren J, Liu L, Li H, Ning Q, Li D, Lu S. The Human Novel Gene LNC-HC Inhibits Hepatocellular Carcinoma Cell Proliferation by Sequestering hsa-miR-183-5p. MOLECULAR THERAPY-NUCLEIC ACIDS 2020; 20:468-479. [PMID: 32278306 PMCID: PMC7150434 DOI: 10.1016/j.omtn.2020.03.008] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 03/17/2020] [Accepted: 03/17/2020] [Indexed: 12/17/2022]
Abstract
Hepatocellular carcinoma (HCC) is the most commonly diagnosed cancer and the leading cause of cancer mortality. Several lines of evidence have demonstrated the aberrant expression of long noncoding RNAs (lncRNAs) in carcinogenesis and their universal regulatory properties. A thorough understanding of lncRNA regulatory roles in HCC pathology would contribute to HCC prevention and treatment. In this study, we identified a novel human lncRNA, LNC-HC, with significantly reduced levels in hepatic tumors from patients with HCC. MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-dimethyltetrazolium bromide) assays as well as colony formation and wound healing experiments showed that LNC-HC significantly inhibited the proliferation of the HCC cell line Huh7. Xenograft transplantation of LNC-HC-overexpressing Huh7 cells in nude mice resulted in the production of smaller tumors. Mechanistically, LNC-HC inhibited the proliferation of HCC cells by directly interacting with hsa-miR-183-5p. LNC-HC rescued the expression of five tumor suppressors, including AKAP12, DYRK2, FOXN3, FOXO1, and LATS2, that were verified as target genes of hsa-miR-183-5p. Overall, human LNC-HC was identified as a novel tumor suppressor that could inhibit HCC cell proliferation in vitro and suppress tumor growth in vivo by competitively binding hsa-miR-183-5p as a competing endogenous RNA (ceRNA). These findings suggest that LNC-HC could be a biomarker of HCC and provide a novel therapeutic target for HCC treatment.
Collapse
Affiliation(s)
- Xi Lan
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China; Key Laboratory of the Environment and Genes Related to Diseases, Xi'an Jiaotong University, Ministry of Education of China, Beijing, China.
| | - Nan Wu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China; Key Laboratory of the Environment and Genes Related to Diseases, Xi'an Jiaotong University, Ministry of Education of China, Beijing, China
| | - Litao Wu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China; Key Laboratory of the Environment and Genes Related to Diseases, Xi'an Jiaotong University, Ministry of Education of China, Beijing, China
| | - Kai Qu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Ezra Kombo Osoro
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China; Key Laboratory of the Environment and Genes Related to Diseases, Xi'an Jiaotong University, Ministry of Education of China, Beijing, China
| | - Dongxian Guan
- Division of Endocrinology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Xiaojuan Du
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China; Key Laboratory of the Environment and Genes Related to Diseases, Xi'an Jiaotong University, Ministry of Education of China, Beijing, China
| | - Bo Wang
- Center for Translational Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Shaanxi 710061, China
| | - Sifan Chen
- Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Ji Miao
- Division of Endocrinology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Juan Ren
- Department of Reproductive Medicine, The Fourth Hospital of Xi'an, Xi'an, Shaanxi 710004, China
| | - Li Liu
- Department of Basic Medical Science, Xi'an Medical College, Xi'an, Shaanxi, China
| | - Haiyun Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China; Key Laboratory of the Environment and Genes Related to Diseases, Xi'an Jiaotong University, Ministry of Education of China, Beijing, China
| | - Qilan Ning
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China; Key Laboratory of the Environment and Genes Related to Diseases, Xi'an Jiaotong University, Ministry of Education of China, Beijing, China
| | - Dongmin Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China; Key Laboratory of the Environment and Genes Related to Diseases, Xi'an Jiaotong University, Ministry of Education of China, Beijing, China
| | - Shemin Lu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China; Key Laboratory of the Environment and Genes Related to Diseases, Xi'an Jiaotong University, Ministry of Education of China, Beijing, China.
| |
Collapse
|
7
|
Konovalova J, Gerasymchuk D, Parkkinen I, Chmielarz P, Domanskyi A. Interplay between MicroRNAs and Oxidative Stress in Neurodegenerative Diseases. Int J Mol Sci 2019; 20:ijms20236055. [PMID: 31801298 PMCID: PMC6929013 DOI: 10.3390/ijms20236055] [Citation(s) in RCA: 135] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 11/23/2019] [Accepted: 11/28/2019] [Indexed: 12/11/2022] Open
Abstract
MicroRNAs are post-transcriptional regulators of gene expression, crucial for neuronal differentiation, survival, and activity. Age-related dysregulation of microRNA biogenesis increases neuronal vulnerability to cellular stress and may contribute to the development and progression of neurodegenerative diseases. All major neurodegenerative disorders are also associated with oxidative stress, which is widely recognized as a potential target for protective therapies. Albeit often considered separately, microRNA networks and oxidative stress are inextricably entwined in neurodegenerative processes. Oxidative stress affects expression levels of multiple microRNAs and, conversely, microRNAs regulate many genes involved in an oxidative stress response. Both oxidative stress and microRNA regulatory networks also influence other processes linked to neurodegeneration, such as mitochondrial dysfunction, deregulation of proteostasis, and increased neuroinflammation, which ultimately lead to neuronal death. Modulating the levels of a relatively small number of microRNAs may therefore alleviate pathological oxidative damage and have neuroprotective activity. Here, we review the role of individual microRNAs in oxidative stress and related pathways in four neurodegenerative conditions: Alzheimer’s (AD), Parkinson’s (PD), Huntington’s (HD) disease, and amyotrophic lateral sclerosis (ALS). We also discuss the problems associated with the use of oversimplified cellular models and highlight perspectives of studying microRNA regulation and oxidative stress in human stem cell-derived neurons.
Collapse
Affiliation(s)
- Julia Konovalova
- Institute of Biotechnology, HiLIFE, University of Helsinki, 00014 Helsinki, Finland; (J.K.); (D.G.); (I.P.)
| | - Dmytro Gerasymchuk
- Institute of Biotechnology, HiLIFE, University of Helsinki, 00014 Helsinki, Finland; (J.K.); (D.G.); (I.P.)
- Institute of Molecular Biology and Genetics, NASU, Kyiv 03143, Ukraine
| | - Ilmari Parkkinen
- Institute of Biotechnology, HiLIFE, University of Helsinki, 00014 Helsinki, Finland; (J.K.); (D.G.); (I.P.)
| | - Piotr Chmielarz
- Department of Brain Biochemistry, Maj Institute of Pharmacology, Polish Academy of Sciences, 31-343 Krakow, Poland
| | - Andrii Domanskyi
- Institute of Biotechnology, HiLIFE, University of Helsinki, 00014 Helsinki, Finland; (J.K.); (D.G.); (I.P.)
- Correspondence: ; Tel.: +358-50-448-4545
| |
Collapse
|
8
|
Chen D, Gong Y, Xu L, Zhou M, Li J, Song J. Bidirectional regulation of osteogenic differentiation by the FOXO subfamily of Forkhead transcription factors in mammalian MSCs. Cell Prolif 2018; 52:e12540. [PMID: 30397974 PMCID: PMC6496202 DOI: 10.1111/cpr.12540] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Revised: 08/09/2018] [Accepted: 09/02/2018] [Indexed: 12/23/2022] Open
Abstract
Through loss‐ and gain‐of‐function experiments in knockout and transgenic mice, Forkhead box O (FOXO) family transcription factors have been demonstrated to play essential roles in many biological processes, including cellular proliferation, apoptosis and differentiation. Osteogenic differentiation from mesenchymal stem cells (MSCs) into osteoblasts is a well‐organized process that is carefully guided and characterized by various factors, such as runt‐related transcription factor 2 (Runx2), β‐catenin, osteocalcin (OCN), alkaline phosphatase (ALP) and activating transcription factor 4 (ATF4). Accumulating evidence suggests multiple interactions among FOXO members and the differentiation regulatory factors listed above, resulting in an enhancement or inhibition of osteogenesis in different stages of osteogenic differentiation. To systematically and integrally understand the role of FOXOs in osteogenic differentiation and explain the contrary phenomena observed in vitro and in vivo, we herein summarized FOXO‐interacting differentiation regulatory genes/factors and following alterations in differentiation. The underlying mechanism was further discussed on the basis of binding types, sites, phases and the consequent downstream transcriptional alterations of interactions among FOXOs and differentiation regulatory factors. Interestingly, a bidirectional effect of FOXOs on balancing osteogenic differentiation was discovered in MSCs. Moreover, FOXO factors are reported to be activated or suppressed by several context‐dependent signalling inputs during differentiation, and the underlying molecular basis may offer new drug development targets for treatments of bone formation defect diseases.
Collapse
Affiliation(s)
- Duanjing Chen
- College of Stomatology, Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, Chongqing, China.,Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Yuanyuan Gong
- College of Stomatology, Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, Chongqing, China.,Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Ling Xu
- College of Stomatology, Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, Chongqing, China.,Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Mengjiao Zhou
- College of Stomatology, Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, Chongqing, China.,Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Jie Li
- College of Stomatology, Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, Chongqing, China.,Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Jinlin Song
- College of Stomatology, Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, Chongqing, China.,Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| |
Collapse
|
9
|
Liu ZQ, Du JJ, Ren JJ, Zhang ZY, Guo XB, Yan YE, Jia XT, Gu NB, Di ZL, Li SZ. miR-183-96-182 clusters alleviated ox-LDL-induced vascular endothelial cell apoptosis in vitro by targeting FOXO1. RSC Adv 2018; 8:35031-35041. [PMID: 35547044 PMCID: PMC9087689 DOI: 10.1039/c8ra06866f] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Accepted: 09/17/2018] [Indexed: 12/13/2022] Open
Abstract
OBJECTIVE To investigate the role of FOXO1 and miR-183-96-182 clusters in ox-LDL induced endothelial cell apoptosis. METHODS FOXO1 overexpression (OE) and knockdown (KD) as well as AKT1 OE in human umbilical vein endothelial cells (HUVECs) and human aortic endothelial cells (HAECs) were achieved by lentiviral transduction. Upregulation of miR-183-5p, miR-182-5p or miR-96-5p was mimicked by agomir treatment. FOXO1 gene transcription was monitored by FOXO1 promotor reporter assay. Cell apoptosis in culture was monitored by TiterTACS in situ detection. Regulation of FOXO1 gene expression by an miRNA targeting mechanism was monitored by AGO2-RNA immunoprecipitation assay. RESULTS FOXO1 mRNA and protein expression levels in ox-LDL treated HUVECs or HAECs were significantly upregulated due to transcriptional and miRNA targeting mechanisms. MiR-183-5p, miR-182-5p and miR-96-5p expression levels in HUVECs or HAECs were significantly reduced by ox-LDL treatment, the overexpression of which by agomir treatment partially reduced the FOXO1 mRNA/protein expression levels and cell apoptosis which was upregulated by ox-LDL treatment. FOXO1 overexpression antagonized the effect of the agomir treatment indicated above. MiR-183-5p, miR-182-5p and miR-96-5p agomir treatment partially rescued the FOXO1 pSer256/total FOXO1 protein ratio and the AKT1 pSer473 level that were reduced by ox-LDL treatment in the HUVECs or HAECs. AKT1 overexpression significantly reduced FOXO1 protein expression, increased miR-182-5p and miR-183-5p expression, and partially alleviated ox-LDL induced HUVEC or HAEC apoptosis in an miR-183-5p and miR-182-5p-dependent manner. CONCLUSION miR-183-96-182 clusters could partially alleviate ox-LDL-induced apoptosis in HUVECs or HAECs by targeting FOXO1.
Collapse
Affiliation(s)
- Zhi-Qin Liu
- Department of Neurology, Xi'an Central Hospital, Xi'an Jiaotong University, School of Medicine Xi'an 710003 Shaanxi China
| | - Jing-Jing Du
- Department of Neurology, Xi'an Central Hospital, Xi'an Jiaotong University, School of Medicine Xi'an 710003 Shaanxi China
| | - Jing-Jing Ren
- Department of Hematology, Xi'an Central Hospital, Xi'an Jiaotong University, School of Medicine Xi'an 710003 Shaanxi China
| | - Zhi-Yong Zhang
- Department of Neurology, China-Japan Friendship Hospital Beijing 100029 China
| | - Xiao-Bo Guo
- Department of Hematology, Xi'an Central Hospital, Xi'an Jiaotong University, School of Medicine Xi'an 710003 Shaanxi China
| | - Yu-E Yan
- Department of Neurology, Xi'an Central Hospital, Xi'an Jiaotong University, School of Medicine Xi'an 710003 Shaanxi China
| | - Xiao-Tao Jia
- Department of Neurology, Xi'an Central Hospital, Xi'an Jiaotong University, School of Medicine Xi'an 710003 Shaanxi China
| | - Nai-Bing Gu
- Department of Neurology, Xi'an Central Hospital, Xi'an Jiaotong University, School of Medicine Xi'an 710003 Shaanxi China
| | - Zheng-Li Di
- Department of Neurology, Xi'an Central Hospital, Xi'an Jiaotong University, School of Medicine Xi'an 710003 Shaanxi China
| | - San-Zhong Li
- Department of Neurosurgery, Xi-jing Hospital Fourth Military Medical University No. 127, Changle Xi Road Xi'an 710032 Shaanxi China +86-185-9140-9510
| |
Collapse
|
10
|
Davis C, Dukes A, Drewry M, Helwa I, Johnson MH, Isales CM, Hill WD, Liu Y, Shi X, Fulzele S, Hamrick MW. MicroRNA-183-5p Increases with Age in Bone-Derived Extracellular Vesicles, Suppresses Bone Marrow Stromal (Stem) Cell Proliferation, and Induces Stem Cell Senescence. Tissue Eng Part A 2017; 23:1231-1240. [PMID: 28363268 PMCID: PMC5689127 DOI: 10.1089/ten.tea.2016.0525] [Citation(s) in RCA: 176] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Accepted: 02/23/2017] [Indexed: 01/08/2023] Open
Abstract
Microvesicle- and exosome-mediated transport of microRNAs (miRNAs) represents a novel cellular and molecular pathway for cell-cell communication. In this study, we tested the hypothesis that these extracellular vesicles (EVs) and their miRNAs might change with age, contributing to age-related stem cell dysfunction. EVs were isolated from the bone marrow interstitial fluid (supernatant) of young (3-4 months) and aged (24-28 months) mice to determine whether the size, concentration, and miRNA profile of EVs were altered with age in vivo. Results show that EVs isolated from bone marrow are CD63 and CD9 positive, and the concentration and size distribution of bone marrow EVs are similar between the young and aged mice. Bioanalyzer data indicate that EVs from both young and aged mice are highly enriched in miRNAs, and the miRNA profile of bone marrow EVs differs significantly between the young and aged mice. Specifically, the miR-183 cluster (miR-96/-182/-183) is highly expressed in aged EVs. In vitro assays demonstrate that aged EVs are endocytosed by primary bone marrow stromal cells (BMSCs), and these aged EVs inhibit the osteogenic differentiation of young BMSCs. Transfection of BMSCs with miR-183-5p mimic reduces cell proliferation and osteogenic differentiation, increases senescence, and decreases protein levels of the miR-183-5p target heme oxygenase-1 (Hmox1). In vitro assays utilizing H2O2-induced oxidative stress show that H2O2 treatment of BMSCs increases the abundance of miR-183-5p in BMSC-derived EVs, and Amplex Red assays demonstrate that H2O2 is elevated in the bone marrow microenvironment with age. Together, these data indicate that aging and oxidative stress can significantly alter the miRNA cargo of EVs in the bone marrow microenvironment, which may in turn play a role in stem cell senescence and osteogenic differentiation by reducing Hmox1 activity.
Collapse
Affiliation(s)
- Colleen Davis
- Department of Cellular Biology & Anatomy, Medical College of Georgia, Augusta University , Augusta, Georgia
| | - Amy Dukes
- Department of Cellular Biology & Anatomy, Medical College of Georgia, Augusta University , Augusta, Georgia
| | - Michelle Drewry
- Department of Cellular Biology & Anatomy, Medical College of Georgia, Augusta University , Augusta, Georgia
| | - Inas Helwa
- Department of Cellular Biology & Anatomy, Medical College of Georgia, Augusta University , Augusta, Georgia
| | - Maribeth H Johnson
- Department of Cellular Biology & Anatomy, Medical College of Georgia, Augusta University , Augusta, Georgia
| | - Carlos M Isales
- Department of Cellular Biology & Anatomy, Medical College of Georgia, Augusta University , Augusta, Georgia
| | - William D Hill
- Department of Cellular Biology & Anatomy, Medical College of Georgia, Augusta University , Augusta, Georgia
| | - Yutao Liu
- Department of Cellular Biology & Anatomy, Medical College of Georgia, Augusta University , Augusta, Georgia
| | - Xingming Shi
- Department of Cellular Biology & Anatomy, Medical College of Georgia, Augusta University , Augusta, Georgia
| | - Sadanand Fulzele
- Department of Cellular Biology & Anatomy, Medical College of Georgia, Augusta University , Augusta, Georgia
| | - Mark W Hamrick
- Department of Cellular Biology & Anatomy, Medical College of Georgia, Augusta University , Augusta, Georgia
| |
Collapse
|
11
|
Liu DZ, Chang B, Li XD, Zhang QH, Zou YH. MicroRNA-9 promotes the proliferation, migration, and invasion of breast cancer cells via down-regulating FOXO1. Clin Transl Oncol 2017; 19:1133-1140. [PMID: 28397066 DOI: 10.1007/s12094-017-1650-1] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2016] [Accepted: 03/13/2017] [Indexed: 02/05/2023]
Abstract
PURPOSE The objective of the study was to investigate the role of microRNA-9 (miR-9) targeting forkhead box O1 (FOXO1) in the proliferation, migration, and invasion of breast cancer cells. METHODS Quantitative real-time polymerase chain reaction (qRT-PCR) was employed to determine the expressions of miR-9 and FOXO1 mRNA in breast cancer tissues, normal breast tissues, breast cancer cell lines, and normal breast epithelial cells. After the up-regulation of miR-9 expression, qRT-PCR and Western blotting were used to determine the expression of FOXO1. The luciferase reporter gene assay was used to validate the target gene. The CCK-8 assay, scratch-wound healing assay, and Transwell invasion assay were used to investigate the changes in the proliferation, migration, and invasion of breast cancer cells, respectively. RESULTS MicroRNA-9 expression was significantly up-regulated in breast cancer tissues and breast cancer cell lines when compared with normal breast tissues and normal breast epithelial cells (both P < 0.05). FOXO1 mRNA and protein expressions were substantially down-regulated in breast cancer tissues and breast cancer cell lines when compared with normal breast tissues and normal breast epithelial cells (both P < 0.05). There can be a negative correlation between miR-9 and FOXO1 mRNA in breast cancer. Luciferase reporter gene assay indicated that miR-9 can down-regulate FOXO1 expression at a post-transcriptional level through binding specifically to FOXO1 3'UTR. The results of CCK-8 assay, scratch-wound healing assay, and Transwell invasion assay revealed that the inhibition of miR-9 can suppress MCF7 cell proliferation, migration, and invasion. Additionally, the expression of miR-9 increased significantly whilst that of FOXO1 decreased substantially as the disease progressed (P < 0.05). CONCLUSIONS Our study provides evidence that miR-9 can promote the proliferation, migration, and invasion of breast cancer cells via down-regulating FOXO1.
Collapse
Affiliation(s)
- D-Z Liu
- Department of Emmengey, The First Affiliated Hospital of Shantou University Medical College, Shantou, 515041, People's Republic of China
| | - B Chang
- Department of Emmengey, The First Affiliated Hospital of Shantou University Medical College, Shantou, 515041, People's Republic of China
| | - X-D Li
- Department of Orthopedics, The First Affiliated Hospital of Shantou University Medical College, Shantou, 515041, People's Republic of China
| | - Q-H Zhang
- Department of Orthopedics, The First Affiliated Hospital of Shantou University Medical College, Shantou, 515041, People's Republic of China
| | - Y-H Zou
- Department of Hepatobiliary Surgery, the First Affiliated Hospital of Shantou University Medical College, No. 57, Changping Road, Shantou, 515041, Guangdong, People's Republic of China.
| |
Collapse
|
12
|
MicroRNAs as regulators and mediators of forkhead box transcription factors function in human cancers. Oncotarget 2017; 8:12433-12450. [PMID: 27999212 PMCID: PMC5355356 DOI: 10.18632/oncotarget.14015] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Accepted: 12/07/2016] [Indexed: 02/07/2023] Open
Abstract
Evidence has shown that microRNAs are widely implicated as indispensable components of tumor suppressive and oncogenic pathways in human cancers. Thus, identification of microRNA targets and their relevant pathways will contribute to the development of microRNA-based therapeutics. The forkhead box transcription factors regulate numerous processes including cell cycle progression, metabolism, metastasis and angiogenesis, thereby facilitating tumor initiation and progression. A complex network of protein and non-coding RNAs mediates the expression and activity of forkhead box transcription factors. In this review, we summarize the current knowledge and concepts concerning the involvement of microRNAs and forkhead box transcription factors and describe the roles of microRNAs-forkhead box axis in various disease states including tumor initiation and progression. Additionally, we describe some of the technical challenges in the use of the microRNA-forkhead box signaling pathway in cancer treatment.
Collapse
|
13
|
Urbánek P, Klotz L. Posttranscriptional regulation of FOXO expression: microRNAs and beyond. Br J Pharmacol 2017; 174:1514-1532. [PMID: 26920226 PMCID: PMC5446586 DOI: 10.1111/bph.13471] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Revised: 02/18/2016] [Accepted: 02/23/2016] [Indexed: 01/17/2023] Open
Abstract
Forkhead box, class O (FOXO) transcription factors are major regulators of diverse cellular processes, including fuel metabolism, oxidative stress response and redox signalling, cell cycle progression and apoptosis. Their activities are controlled by multiple posttranslational modifications and nuclear-cytoplasmic shuttling. Recently, post-transcriptional regulation of FOXO synthesis has emerged as a new regulatory level of their functions. Accumulating evidence suggests that this post-transcriptional mode of regulation of FOXO activity operates in response to stressful stimuli, including oxidative stress. Here, we give a brief overview on post-transcriptional regulation of FOXO synthesis by microRNAs (miRNAs) and by RNA-binding regulatory proteins, human antigen R (HuR) and quaking (QKI). Aberrant post-transcriptional regulation of FOXOs is frequently connected with various disease states. We therefore discuss characteristic examples of FOXO regulation at the post-transcriptional level under various physiological and pathophysiological conditions, including oxidative stress and cancer. The picture emerging from this summary points to a diversity of interactions between miRNAs/miRNA-induced silencing complexes and RNA-binding regulatory proteins. Better insight into these complexities of post-transcriptional regulatory interactions will add to our understanding of the mechanisms of pathological processes and the role of FOXO proteins. LINKED ARTICLES This article is part of a themed section on Redox Biology and Oxidative Stress in Health and Disease. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v174.12/issuetoc.
Collapse
Affiliation(s)
- P Urbánek
- Institute of Nutrition, Department of NutrigenomicsFriedrich‐Schiller‐Universität JenaJenaGermany
| | - L‐O Klotz
- Institute of Nutrition, Department of NutrigenomicsFriedrich‐Schiller‐Universität JenaJenaGermany
| |
Collapse
|
14
|
França GS, Hinske LC, Galante PAF, Vibranovski MD. Unveiling the Impact of the Genomic Architecture on the Evolution of Vertebrate microRNAs. Front Genet 2017; 8:34. [PMID: 28377786 PMCID: PMC5359303 DOI: 10.3389/fgene.2017.00034] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2016] [Accepted: 03/09/2017] [Indexed: 12/12/2022] Open
Abstract
Eukaryotic genomes frequently exhibit interdependency between transcriptional units, as evidenced by regions of high gene density. It is well recognized that vertebrate microRNAs (miRNAs) are usually embedded in those regions. Recent work has shown that the genomic context is of utmost importance to determine miRNA expression in time and space, thus affecting their evolutionary fates over long and short terms. Consequently, understanding the inter- and intraspecific changes on miRNA genomic architecture may bring novel insights on the basic cellular processes regulated by miRNAs, as well as phenotypic evolution and disease-related mechanisms.
Collapse
Affiliation(s)
- Gustavo S França
- Departamento de Genética e Biologia Evolutiva, Universidade de São Paulo São Paulo, Brazil
| | - Ludwig C Hinske
- Department of Anesthesiology, Clinic of the University of Munich, Ludwig Maximilian University of Munich Munich, Germany
| | - Pedro A F Galante
- Centro de Oncologia Molecular, Hospital Sírio-Libanês São Paulo, Brazil
| | - Maria D Vibranovski
- Departamento de Genética e Biologia Evolutiva, Universidade de São Paulo São Paulo, Brazil
| |
Collapse
|
15
|
Wang Y, Veremeyko T, Wong AHK, El Fatimy R, Wei Z, Cai W, Krichevsky AM. Downregulation of miR-132/212 impairs S-nitrosylation balance and induces tau phosphorylation in Alzheimer's disease. Neurobiol Aging 2016; 51:156-166. [PMID: 28089352 DOI: 10.1016/j.neurobiolaging.2016.12.015] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Revised: 12/16/2016] [Accepted: 12/18/2016] [Indexed: 01/31/2023]
Abstract
MicroRNA-132 is markedly downregulated in Alzheimer's disease (AD) and related tauopathies, and its levels are closely associated with tau pathology in AD. Whether and how miR-132 contributes to pathology in these neurodegenerative diseases remains unclear. Here, we show that miR-132 and its paralogue miR-212 directly regulate the expression of neuronal nitric oxide synthase (NOS1) through the primate-specific binding site. Inhibition of miR-132 in primary human neurons and neural cells leads to increased NOS1 levels and triggers excessive production of nitric oxide, followed by aberrant S-nitrosylation (SNO) of specific proteins associated with neurodegeneration and tau pathology, such as cyclin-dependent kinase 5, dynamin-related protein 1, and glyceraldehyde-3-phosphate dehydrogenase. This, in turn, increases tau phosphorylation at disease associated Ser396, Ser404, and Ser202 sites, and impairs neural viability. Our findings indicate that downregulation of miR-132/212 disturbs the balance of S-nitrosylation and induces tau phosphorylation in a NOS1-dependent way, and thereby may contribute to the pathogenesis of AD and other tauopathies.
Collapse
Affiliation(s)
- Yang Wang
- Department of Pediatric Surgery, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China; Department of Neurology, Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Tatiana Veremeyko
- Department of Neurology, Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Andus Hon-Kit Wong
- Department of Neurology, Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Rachid El Fatimy
- Department of Neurology, Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Zhiyun Wei
- Department of Neurology, Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Wei Cai
- Department of Pediatric Surgery, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Anna M Krichevsky
- Department of Neurology, Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
16
|
Kim BK, Kim I, Lee AR, Yoo HI, Yoon SK. Mouse-specific up-regulation of Ccnb1 expression by miR-199a-5p in keratinocyte. FEBS Open Bio 2016; 6:1131-1140. [PMID: 27833853 PMCID: PMC5095150 DOI: 10.1002/2211-5463.12133] [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: 05/05/2016] [Revised: 08/10/2016] [Accepted: 09/04/2016] [Indexed: 01/07/2023] Open
Abstract
MicroRNA (miRNA) are a class of single-stranded, small non-coding RNA that regulate various biological processes, including skin and hair cycle regulation, by modulating the expression of specific genes at the post-transcriptional level. Recently, several studies reported that miRNA directly or indirectly up-regulate target genes. Previously, we performed microarray analysis to identify the target genes of miR-199a-5p in a mouse skin keratinocyte cell line and detected more than 200 genes whose expression was significantly increased by miR-199a-5p overexpression (> 1.5-fold). In this study, we further investigated these genes and found that cyclin B1 (Ccnb1) expression was positively regulated by miR-199a-5p in keratinocyte. Moreover, Ccnb1 expression was inversely correlated with miR-199a-5p expression during the mouse hair cycle. Cell cycle analysis showed that the proportion of cells in S phase was slightly increased, while the proportion of cells in G2/M phase decreased by miR-199-5p. Using luciferase assay, we found that the 3' untranslated region of Ccnb1 was a direct target of miR-199a-5p. We also found that the regulation of Ccnb1 expression by miR-199a-5p is mouse specific. CCNB1 expression was not affected in the human and monkey cell lines. These results provide a new relationship between Ccnb1 and miR-199a-5p in both mouse keratinocyte and miRNA biology.
Collapse
Affiliation(s)
- Bong-Kyu Kim
- Department of Medical Lifesciences The Catholic University of Korea Seoul Korea
| | - Injung Kim
- Department of Medical Lifesciences The Catholic University of Korea Seoul Korea
| | - Ah-Reum Lee
- Department of Medical Lifesciences The Catholic University of Korea Seoul Korea
| | - Hye-In Yoo
- Department of Medical Lifesciences The Catholic University of Korea Seoul Korea
| | - Sungjoo Kim Yoon
- Department of Medical Lifesciences The Catholic University of Korea Seoul Korea
| |
Collapse
|
17
|
Perdomo-Sabogal A, Kanton S, Walter MBC, Nowick K. The role of gene regulatory factors in the evolutionary history of humans. Curr Opin Genet Dev 2014; 29:60-7. [PMID: 25215414 DOI: 10.1016/j.gde.2014.08.007] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2014] [Revised: 08/05/2014] [Accepted: 08/22/2014] [Indexed: 01/01/2023]
Abstract
Deciphering the molecular basis of how modern human phenotypes have evolved is one of the most fascinating challenges in biology. Here, we will focus on the roles of gene regulatory factors (GRFs), in particular transcription factors (TFs) and long non-coding RNAs (lncRNAs) during human evolution. We will present examples of TFs and lncRNAs that have changed or show signs of positive selection in humans compared to chimpanzees, in modern humans compared to archaic humans, or within modern human populations. On the basis of current knowledge about the functions of these GRF genes, we speculate that they have been involved in speciation as well as in shaping phenotypes such as brain functions, skeletal morphology, and metabolic processes.
Collapse
Affiliation(s)
- Alvaro Perdomo-Sabogal
- TFome Research Group, Bioinformatics Group, Interdisciplinary Center of Bioinformatics, Department of Computer Science, University of Leipzig, Härtelstrasse 16-18, D-04107 Leipzig, Germany; Paul-Flechsig-Institute for Brain Research, University of Leipzig, Jahnallee 59, D-04109 Leipzig, Germany
| | - Sabina Kanton
- TFome Research Group, Bioinformatics Group, Interdisciplinary Center of Bioinformatics, Department of Computer Science, University of Leipzig, Härtelstrasse 16-18, D-04107 Leipzig, Germany; Paul-Flechsig-Institute for Brain Research, University of Leipzig, Jahnallee 59, D-04109 Leipzig, Germany
| | - Maria Beatriz C Walter
- TFome Research Group, Bioinformatics Group, Interdisciplinary Center of Bioinformatics, Department of Computer Science, University of Leipzig, Härtelstrasse 16-18, D-04107 Leipzig, Germany; Paul-Flechsig-Institute for Brain Research, University of Leipzig, Jahnallee 59, D-04109 Leipzig, Germany
| | - Katja Nowick
- TFome Research Group, Bioinformatics Group, Interdisciplinary Center of Bioinformatics, Department of Computer Science, University of Leipzig, Härtelstrasse 16-18, D-04107 Leipzig, Germany; Paul-Flechsig-Institute for Brain Research, University of Leipzig, Jahnallee 59, D-04109 Leipzig, Germany.
| |
Collapse
|