151
|
Zhang XM, Guo L, Chi MH, Sun HM, Chen XW. Identification of active miRNA and transcription factor regulatory pathways in human obesity-related inflammation. BMC Bioinformatics 2015; 16:76. [PMID: 25887648 PMCID: PMC4355475 DOI: 10.1186/s12859-015-0512-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2014] [Accepted: 02/24/2015] [Indexed: 12/21/2022] Open
Abstract
Background Obesity-induced chronic inflammation plays a fundamental role in the pathogenesis of metabolic syndrome (MS). Recently, a growing body of evidence supports that miRNAs are largely dysregulated in obesity and that specific miRNAs regulate obesity-associated inflammation. We applied an approach aiming to identify active miRNA-TF-gene regulatory pathways in obesity. Firstly, we detected differentially expressed genes (DEGs) and differentially expressed miRNAs (DEmiRs) from mRNA and miRNA expression profiles, respectively. Secondly, by mapping the DEGs and DEmiRs to the curated miRNA-TF-gene regulatory network as active seed nodes and connect them with their immediate neighbors, we obtained the potential active miRNA-TF-gene regulatory subnetwork in obesity. Thirdly, using a Breadth-First-Search (BFS) algorithm, we identified potential active miRNA-TF-gene regulatory pathways in obesity. Finally, through the hypergeometric test, we identified the active miRNA-TF-gene regulatory pathways that were significantly related to obesity. Results The potential active pathways with FDR < 0.0005 were considered to be the active miRNA-TF regulatory pathways in obesity. The union of the active pathways is visualized and identical nodes of the active pathways were merged. Conclusions We identified 23 active miRNA-TF-gene regulatory pathways that were significantly related to obesity-related inflammation. Electronic supplementary material The online version of this article (doi:10.1186/s12859-015-0512-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Xi-Mei Zhang
- Department of Histology and Embryology, Harbin Medical University, Harbin, 150081, PR China.
| | - Lin Guo
- Department of Endocrinology and Metabolism, the Second Affiliated Hospital of Harbin Medical University, Harbin, 150081, PR China.
| | - Mei-Hua Chi
- Teaching Experiment Center of Morphology, Harbin Medical University, Harbin, 150081, PR China.
| | - Hong-Mei Sun
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, PR China.
| | - Xiao-Wen Chen
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, PR China.
| |
Collapse
|
152
|
Vengrenyuk Y, Nishi H, Long X, Ouimet M, Savji N, Martinez FO, Cassella CP, Moore KJ, Ramsey SA, Miano JM, Fisher EA. Cholesterol loading reprograms the microRNA-143/145-myocardin axis to convert aortic smooth muscle cells to a dysfunctional macrophage-like phenotype. Arterioscler Thromb Vasc Biol 2015; 35:535-46. [PMID: 25573853 PMCID: PMC4344402 DOI: 10.1161/atvbaha.114.304029] [Citation(s) in RCA: 279] [Impact Index Per Article: 27.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
OBJECTIVE We previously showed that cholesterol loading in vitro converts mouse aortic vascular smooth muscle cells (VSMC) from a contractile state to one resembling macrophages. In human and mouse atherosclerotic plaques, it has become appreciated that ≈40% of cells classified as macrophages by histological markers may be of VSMC origin. Therefore, we sought to gain insight into the molecular regulation of this clinically relevant process. APPROACH AND RESULTS VSMC of mouse (or human) origin were incubated with cyclodextrin-cholesterol complexes for 72 hours, at which time the expression at the protein and mRNA levels of contractile-related proteins was reduced and of macrophage markers increased. Concurrent was downregulation of miR-143/145, which positively regulate the master VSMC differentiation transcription factor myocardin. Mechanisms were further probed in mouse VSMC. Maintaining the expression of myocardin or miR-143/145 prevented and reversed phenotypic changes caused by cholesterol loading. Reversal was also seen when cholesterol efflux was stimulated after loading. Notably, despite expression of macrophage markers, bioinformatic analyses showed that cholesterol-loaded cells remained closer to the VSMC state, consistent with impairment in classical macrophage functions of phagocytosis and efferocytosis. In apoE-deficient atherosclerotic plaques, cells positive for VSMC and macrophage markers were found lining the cholesterol-rich necrotic core. CONCLUSIONS Cholesterol loading of VSMC converts them to a macrophage-appearing state by downregulating the miR-143/145-myocardin axis. Although these cells would be classified by immunohistochemistry as macrophages in human and mouse plaques, their transcriptome and functional properties imply that their contributions to atherogenesis would not be those of classical macrophages.
Collapse
MESH Headings
- Animals
- Aorta, Thoracic/metabolism
- Aorta, Thoracic/pathology
- Apolipoproteins E/deficiency
- Apolipoproteins E/genetics
- Atherosclerosis/genetics
- Atherosclerosis/metabolism
- Atherosclerosis/pathology
- Binding Sites
- Cell Lineage
- Cell Transdifferentiation
- Cholesterol/metabolism
- Cholesterol, HDL/metabolism
- Coculture Techniques
- Disease Models, Animal
- Foam Cells/metabolism
- Foam Cells/pathology
- Gene Expression Profiling/methods
- Gene Expression Regulation
- Humans
- Jurkat Cells
- Mice, Inbred C57BL
- Mice, Knockout
- MicroRNAs/genetics
- MicroRNAs/metabolism
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Necrosis
- Nuclear Proteins/genetics
- Nuclear Proteins/metabolism
- Oligonucleotide Array Sequence Analysis
- Phagocytosis
- Phenotype
- Plaque, Atherosclerotic
- Signal Transduction
- Sterol Regulatory Element Binding Protein 2/metabolism
- Time Factors
- Trans-Activators/genetics
- Trans-Activators/metabolism
- Transfection
Collapse
Affiliation(s)
- Yuliya Vengrenyuk
- From the Marc and Ruti Bell Program in Vascular Biology, Division of Cardiology, Department of Medicine, NYU School of Medicine, New York (Y.V., H.N., M.O., N.S., C.P.C., K.J.M., E.A.F.); Center for Cardiovascular Sciences, Albany Medical College, NY (X.L.); Nuffield Department of Orthopaedics, Rheumatology, and Musculoskeletal Sciences, Botnar Research Centre, University of Oxford, Oxford, United Kingdom (F.O.M.); Department of Biomedical Sciences, Oregon State University, Corvallis (S.A.R.); and Department of Medicine, Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, NY (J.M.M.)
| | - Hitoo Nishi
- From the Marc and Ruti Bell Program in Vascular Biology, Division of Cardiology, Department of Medicine, NYU School of Medicine, New York (Y.V., H.N., M.O., N.S., C.P.C., K.J.M., E.A.F.); Center for Cardiovascular Sciences, Albany Medical College, NY (X.L.); Nuffield Department of Orthopaedics, Rheumatology, and Musculoskeletal Sciences, Botnar Research Centre, University of Oxford, Oxford, United Kingdom (F.O.M.); Department of Biomedical Sciences, Oregon State University, Corvallis (S.A.R.); and Department of Medicine, Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, NY (J.M.M.)
| | - Xiaochun Long
- From the Marc and Ruti Bell Program in Vascular Biology, Division of Cardiology, Department of Medicine, NYU School of Medicine, New York (Y.V., H.N., M.O., N.S., C.P.C., K.J.M., E.A.F.); Center for Cardiovascular Sciences, Albany Medical College, NY (X.L.); Nuffield Department of Orthopaedics, Rheumatology, and Musculoskeletal Sciences, Botnar Research Centre, University of Oxford, Oxford, United Kingdom (F.O.M.); Department of Biomedical Sciences, Oregon State University, Corvallis (S.A.R.); and Department of Medicine, Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, NY (J.M.M.)
| | - Mireille Ouimet
- From the Marc and Ruti Bell Program in Vascular Biology, Division of Cardiology, Department of Medicine, NYU School of Medicine, New York (Y.V., H.N., M.O., N.S., C.P.C., K.J.M., E.A.F.); Center for Cardiovascular Sciences, Albany Medical College, NY (X.L.); Nuffield Department of Orthopaedics, Rheumatology, and Musculoskeletal Sciences, Botnar Research Centre, University of Oxford, Oxford, United Kingdom (F.O.M.); Department of Biomedical Sciences, Oregon State University, Corvallis (S.A.R.); and Department of Medicine, Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, NY (J.M.M.)
| | - Nazir Savji
- From the Marc and Ruti Bell Program in Vascular Biology, Division of Cardiology, Department of Medicine, NYU School of Medicine, New York (Y.V., H.N., M.O., N.S., C.P.C., K.J.M., E.A.F.); Center for Cardiovascular Sciences, Albany Medical College, NY (X.L.); Nuffield Department of Orthopaedics, Rheumatology, and Musculoskeletal Sciences, Botnar Research Centre, University of Oxford, Oxford, United Kingdom (F.O.M.); Department of Biomedical Sciences, Oregon State University, Corvallis (S.A.R.); and Department of Medicine, Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, NY (J.M.M.)
| | - Fernando O Martinez
- From the Marc and Ruti Bell Program in Vascular Biology, Division of Cardiology, Department of Medicine, NYU School of Medicine, New York (Y.V., H.N., M.O., N.S., C.P.C., K.J.M., E.A.F.); Center for Cardiovascular Sciences, Albany Medical College, NY (X.L.); Nuffield Department of Orthopaedics, Rheumatology, and Musculoskeletal Sciences, Botnar Research Centre, University of Oxford, Oxford, United Kingdom (F.O.M.); Department of Biomedical Sciences, Oregon State University, Corvallis (S.A.R.); and Department of Medicine, Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, NY (J.M.M.)
| | - Courtney P Cassella
- From the Marc and Ruti Bell Program in Vascular Biology, Division of Cardiology, Department of Medicine, NYU School of Medicine, New York (Y.V., H.N., M.O., N.S., C.P.C., K.J.M., E.A.F.); Center for Cardiovascular Sciences, Albany Medical College, NY (X.L.); Nuffield Department of Orthopaedics, Rheumatology, and Musculoskeletal Sciences, Botnar Research Centre, University of Oxford, Oxford, United Kingdom (F.O.M.); Department of Biomedical Sciences, Oregon State University, Corvallis (S.A.R.); and Department of Medicine, Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, NY (J.M.M.)
| | - Kathryn J Moore
- From the Marc and Ruti Bell Program in Vascular Biology, Division of Cardiology, Department of Medicine, NYU School of Medicine, New York (Y.V., H.N., M.O., N.S., C.P.C., K.J.M., E.A.F.); Center for Cardiovascular Sciences, Albany Medical College, NY (X.L.); Nuffield Department of Orthopaedics, Rheumatology, and Musculoskeletal Sciences, Botnar Research Centre, University of Oxford, Oxford, United Kingdom (F.O.M.); Department of Biomedical Sciences, Oregon State University, Corvallis (S.A.R.); and Department of Medicine, Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, NY (J.M.M.)
| | - Stephen A Ramsey
- From the Marc and Ruti Bell Program in Vascular Biology, Division of Cardiology, Department of Medicine, NYU School of Medicine, New York (Y.V., H.N., M.O., N.S., C.P.C., K.J.M., E.A.F.); Center for Cardiovascular Sciences, Albany Medical College, NY (X.L.); Nuffield Department of Orthopaedics, Rheumatology, and Musculoskeletal Sciences, Botnar Research Centre, University of Oxford, Oxford, United Kingdom (F.O.M.); Department of Biomedical Sciences, Oregon State University, Corvallis (S.A.R.); and Department of Medicine, Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, NY (J.M.M.)
| | - Joseph M Miano
- From the Marc and Ruti Bell Program in Vascular Biology, Division of Cardiology, Department of Medicine, NYU School of Medicine, New York (Y.V., H.N., M.O., N.S., C.P.C., K.J.M., E.A.F.); Center for Cardiovascular Sciences, Albany Medical College, NY (X.L.); Nuffield Department of Orthopaedics, Rheumatology, and Musculoskeletal Sciences, Botnar Research Centre, University of Oxford, Oxford, United Kingdom (F.O.M.); Department of Biomedical Sciences, Oregon State University, Corvallis (S.A.R.); and Department of Medicine, Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, NY (J.M.M.)
| | - Edward A Fisher
- From the Marc and Ruti Bell Program in Vascular Biology, Division of Cardiology, Department of Medicine, NYU School of Medicine, New York (Y.V., H.N., M.O., N.S., C.P.C., K.J.M., E.A.F.); Center for Cardiovascular Sciences, Albany Medical College, NY (X.L.); Nuffield Department of Orthopaedics, Rheumatology, and Musculoskeletal Sciences, Botnar Research Centre, University of Oxford, Oxford, United Kingdom (F.O.M.); Department of Biomedical Sciences, Oregon State University, Corvallis (S.A.R.); and Department of Medicine, Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, NY (J.M.M.).
| |
Collapse
|
153
|
Huang JY, Chou SF, Lee JW, Chen HL, Chen CM, Tao MH, Shih C. MicroRNA-130a can inhibit hepatitis B virus replication via targeting PGC1α and PPARγ. RNA (NEW YORK, N.Y.) 2015; 21:385-400. [PMID: 25595716 PMCID: PMC4338335 DOI: 10.1261/rna.048744.114] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/25/2014] [Accepted: 12/08/2014] [Indexed: 06/04/2023]
Abstract
In hepatitis B virus (HBV)-replicating hepatocytes, miR-130a expression was significantly reduced. In a reciprocal manner, miR-130a reduced HBV replication by targeting at two major metabolic regulators PGC1α and PPARγ, both of which can potently stimulate HBV replication. We proposed a positive feed-forward loop between HBV, miR-130a, PPARγ, and PGC1α. Accordingly, HBV can significantly enhance viral replication by reducing miR-130a and increasing PGC1α and PPARγ. NF-κB/p65 can strongly stimulate miR-130a promoter, while miR-130a can promote NF-κB/p65 protein level by reducing PPARγ and thus NF-κB/p65 protein degradation. We postulated another positive feed-forward loop between miR-130a and NF-κB/p65 via PPARγ. During liver inflammation, NF-κB signaling could contribute to viral clearance via its positive effect on miR-130a transcription. Conversely, in asymptomatic HBV carriers, persistent viral infection could reduce miR-130a and NF-κB expression, leading to dampened inflammation and immune tolerance. Finally, miR-130a could contribute to metabolic homeostasis by dual targeting PGC1α and PPARγ simultaneously.
Collapse
Affiliation(s)
- Jyun-Yuan Huang
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, 114 Taiwan Institute of Biomedical Sciences, Academia Sinica, Taipei, 115 Taiwan
| | - Shu-Fan Chou
- Institute of Biomedical Sciences, Academia Sinica, Taipei, 115 Taiwan Graduate Institute of Microbiology, College of Medicine, National Taiwan University, Taipei, 110 Taiwan
| | - Jun-Wei Lee
- Institute of Biomedical Sciences, Academia Sinica, Taipei, 115 Taiwan
| | - Hung-Lin Chen
- Institute of Biomedical Sciences, Academia Sinica, Taipei, 115 Taiwan
| | - Chun-Ming Chen
- Institute of Biomedical Sciences, Academia Sinica, Taipei, 115 Taiwan
| | - Mi-Hua Tao
- Institute of Biomedical Sciences, Academia Sinica, Taipei, 115 Taiwan
| | - Chiaho Shih
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, 114 Taiwan Institute of Biomedical Sciences, Academia Sinica, Taipei, 115 Taiwan
| |
Collapse
|
154
|
Muehlschlegel JD, Christodoulou DC, McKean D, Gorham J, Mazaika E, Heydarpour M, Lee G, DePalma SR, Perry TE, Fox AA, Shernan SK, Seidman CE, Aranki SF, Seidman JG, Body SC. Using next-generation RNA sequencing to examine ischemic changes induced by cold blood cardioplegia on the human left ventricular myocardium transcriptome. Anesthesiology 2015; 122:537-50. [PMID: 25581909 PMCID: PMC4333028 DOI: 10.1097/aln.0000000000000582] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
BACKGROUND The exact mechanisms that underlie the pathological processes of myocardial ischemia in humans are unclear. Cardiopulmonary bypass with cardioplegic arrest allows the authors to examine the whole transcriptional profile of human left ventricular myocardium at baseline and after exposure to cold cardioplegia-induced ischemia as a human ischemia model. METHODS The authors obtained biopsies from 45 patients undergoing aortic valve replacement surgery at baseline and after an average of 79 min of cold cardioplegic arrest. Samples were RNA sequenced and analyzed with the Partek Genomics Suite (Partek Inc., St. Louis, MO) for differential expression. Ingenuity Pathway Analysis (Ingenuity Systems, Redwood City, CA) and Biobase ExPlain (Biobase GmbH, Wolfenbuettel, Germany) systems were used for functional and pathway analyses. RESULTS Of the 4,098 genes with a mean expression value greater than 5, 90% were down-regulated and 9.1% were up-regulated. Of those, 1,241 were significantly differentially expressed. Gene ontology analysis revealed significant down-regulation in immune inflammatory response and complement activation categories and highly consistent was the down-regulation of intelectin 1, proteoglycan, and secretory leukocyte peptidase inhibitor. Up-regulated genes of interest were FBJ murine osteosarcoma viral oncogene homolog and the hemoglobin genes hemoglobin α1 (HBA1) and hemoglobin β. In addition, analysis of transcription factor-binding sites revealed interesting targets in factors regulating reactive oxygen species production, apoptosis, immunity, cytokine production, and inflammatory response. CONCLUSIONS The authors have shown that the human left ventricle exhibits significant changes in gene expression in response to cold cardioplegia-induced ischemia during cardiopulmonary bypass, which provides great insight into the pathophysiology of ventricular ischemia, and thus, may help guide efforts to reduce myocardial damage during surgery.
Collapse
Affiliation(s)
- Jochen D Muehlschlegel
- From the Brigham and Women's Hospital, Department of Anesthesiology, Perioperative and Pain Medicine, Harvard Medical School, Boston, Massachusetts (J.D.M., M.H., S.K.S., S.C.B.); Department of Genetics, Harvard Medical School, Boston, Massachusetts (D.C.C., D.M., J.G., E.M., S.R.D., J.G.S.); Harvard Medical School, Boston, Massachusetts (G.L.); Northwest Anesthesia, Abbott Northwestern Hospital and Minneapolis Heart Institute, Minneapolis, Minnesota (T.E.P.); Department of Anesthesiology, University of Texas Southwestern Medical Center, Dallas, Texas (A.A.F.); Howard Hughes Medical Institute, Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts (C.E.S.); and Brigham and Women's Hospital, Division of Cardiac Surgery, Department of Surgery, Harvard Medical School, Boston, Massachusetts (S.F.A.)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
155
|
Bajaj D, Saxena MS, Kujur A, Das S, Badoni S, Tripathi S, Upadhyaya HD, Gowda CLL, Sharma S, Singh S, Tyagi AK, Parida SK. Genome-wide conserved non-coding microsatellite (CNMS) marker-based integrative genetical genomics for quantitative dissection of seed weight in chickpea. JOURNAL OF EXPERIMENTAL BOTANY 2015; 66:1271-90. [PMID: 25504138 PMCID: PMC4339591 DOI: 10.1093/jxb/eru478] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2023]
Abstract
Phylogenetic footprinting identified 666 genome-wide paralogous and orthologous CNMS (conserved non-coding microsatellite) markers from 5'-untranslated and regulatory regions (URRs) of 603 protein-coding chickpea genes. The (CT)n and (GA)n CNMS carrying CTRMCAMV35S and GAGA8BKN3 regulatory elements, respectively, are abundant in the chickpea genome. The mapped genic CNMS markers with robust amplification efficiencies (94.7%) detected higher intraspecific polymorphic potential (37.6%) among genotypes, implying their immense utility in chickpea breeding and genetic analyses. Seventeen differentially expressed CNMS marker-associated genes showing strong preferential and seed tissue/developmental stage-specific expression in contrasting genotypes were selected to narrow down the gene targets underlying seed weight quantitative trait loci (QTLs)/eQTLs (expression QTLs) through integrative genetical genomics. The integration of transcript profiling with seed weight QTL/eQTL mapping, molecular haplotyping, and association analyses identified potential molecular tags (GAGA8BKN3 and RAV1AAT regulatory elements and alleles/haplotypes) in the LOB-domain-containing protein- and KANADI protein-encoding transcription factor genes controlling the cis-regulated expression for seed weight in the chickpea. This emphasizes the potential of CNMS marker-based integrative genetical genomics for the quantitative genetic dissection of complex seed weight in chickpea.
Collapse
Affiliation(s)
- Deepak Bajaj
- National Institute of Plant Genome Research (NIPGR), Aruna Asaf Ali Marg, New Delhi 110067, India
| | - Maneesha S Saxena
- National Institute of Plant Genome Research (NIPGR), Aruna Asaf Ali Marg, New Delhi 110067, India
| | - Alice Kujur
- National Institute of Plant Genome Research (NIPGR), Aruna Asaf Ali Marg, New Delhi 110067, India
| | - Shouvik Das
- National Institute of Plant Genome Research (NIPGR), Aruna Asaf Ali Marg, New Delhi 110067, India
| | - Saurabh Badoni
- National Institute of Plant Genome Research (NIPGR), Aruna Asaf Ali Marg, New Delhi 110067, India
| | - Shailesh Tripathi
- Division of Genetics, Indian Agricultural Research Institute (IARI), New Delhi 110012, India
| | - Hari D Upadhyaya
- International Crops Research Institute for the Semi-Arid Tropics (ICRISAT), Patancheru 502324, Telangana, India
| | - C L L Gowda
- International Crops Research Institute for the Semi-Arid Tropics (ICRISAT), Patancheru 502324, Telangana, India
| | - Shivali Sharma
- International Crops Research Institute for the Semi-Arid Tropics (ICRISAT), Patancheru 502324, Telangana, India
| | - Sube Singh
- International Crops Research Institute for the Semi-Arid Tropics (ICRISAT), Patancheru 502324, Telangana, India
| | - Akhilesh K Tyagi
- National Institute of Plant Genome Research (NIPGR), Aruna Asaf Ali Marg, New Delhi 110067, India
| | - Swarup K Parida
- National Institute of Plant Genome Research (NIPGR), Aruna Asaf Ali Marg, New Delhi 110067, India
| |
Collapse
|
156
|
Rothenberg SM, Concannon K, Cullen S, Boulay G, Turke AB, Faber AC, Lockerman EL, Rivera MN, Engelman JA, Maheswaran S, Haber DA. Inhibition of mutant EGFR in lung cancer cells triggers SOX2-FOXO6-dependent survival pathways. eLife 2015; 4. [PMID: 25686219 PMCID: PMC4384750 DOI: 10.7554/elife.06132] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Accepted: 02/03/2015] [Indexed: 12/17/2022] Open
Abstract
Treatment of EGFR-mutant lung cancer with erlotinib results in
dramatic tumor regression but it is invariably followed by drug resistance. In
characterizing early transcriptional changes following drug treatment of mutant
EGFR-addicted cells, we identified the stem cell transcriptional regulator SOX2 as
being rapidly and specifically induced, both in vitro and in vivo. Suppression of
SOX2 sensitizes cells to erlotinib-mediated apoptosis, ultimately decreasing the
emergence of acquired resistance, whereas its ectopic expression reduces drug-induced
cell death. We show that erlotinib relieves EGFR-dependent suppression of FOXO6,
leading to its induction of SOX2, which in turn represses the pro-apoptotic BH3-only
genes BIM and BMF. Together, these observations
point to a physiological feedback mechanism that attenuates oncogene
addiction-mediated cell death associated with the withdrawal of growth factor
signaling and may therefore contribute to the development of resistance. DOI:http://dx.doi.org/10.7554/eLife.06132.001 Tumors can form when cells gain mutations in genes that enable them to grow and
divide rapidly. In some human lung cancers, genetic mutations are found in a gene
that makes a protein called EGFR. This protein encourages cells to divide and the
mutations can lead to the cancer cells producing more EGFR, or producing a form of
the protein that is more active. Treating these cancers with a drug called erlotinib inhibits EGFR and makes the
tumors shrink dramatically, but the tumors will usually re-grow because any tumor
cells that survive often become resistant to the drug. There are several ways that
the tumor cells can become resistant, which makes the task of developing a solution
to this problem more difficult. It has been suggested that the tumor cells may enter
a temporary ‘drug-tolerant’ state that helps them to survive and makes
it more likely that they will develop resistance to the drug. However, it is not
clear how this drug-tolerant state might work. To address this question, Rothenberg et al. examined which genes are switched on (or
‘expressed’) in tumor cells with a mutant version of EGFR after they
were treated with the erlotinib drug. The experiments show that a gene called
SOX2 is expressed in these cells. Cells that had lower levels of
SOX2 expression were more sensitive to the effects of the drug
and fewer cells developed resistance. On the other hand, cells that had higher levels
of SOX2 expression were less sensitive to the drug and resistance
was more likely to develop. A protein called FOXO6—which is usually suppressed by EGFR—activates
the SOX2 gene in these cells. Therefore, using erlotinib to inhibit
EGFR to kill the cancer cells increases the activity of FOXO6, which in turn promotes
the survival of some of the cells by activating the SOX2 gene. A
better understanding of the ways in which cancer cells adapt to erlotinib and other
drugs may help us to design more effective treatments with better outcomes for
patients. DOI:http://dx.doi.org/10.7554/eLife.06132.002
Collapse
Affiliation(s)
- S Michael Rothenberg
- Cancer Center, Massachusetts General Hospital, Harvard Medical School, Charlestown, United States
| | - Kyle Concannon
- Cancer Center, Massachusetts General Hospital, Harvard Medical School, Charlestown, United States
| | - Sarah Cullen
- Cancer Center, Massachusetts General Hospital, Harvard Medical School, Charlestown, United States
| | - Gaylor Boulay
- Cancer Center, Massachusetts General Hospital, Harvard Medical School, Charlestown, United States
| | - Alexa B Turke
- Cancer Center, Massachusetts General Hospital, Harvard Medical School, Charlestown, United States
| | - Anthony C Faber
- Cancer Center, Massachusetts General Hospital, Harvard Medical School, Charlestown, United States
| | - Elizabeth L Lockerman
- Cancer Center, Massachusetts General Hospital, Harvard Medical School, Charlestown, United States
| | - Miguel N Rivera
- Cancer Center, Massachusetts General Hospital, Harvard Medical School, Charlestown, United States
| | - Jeffrey A Engelman
- Cancer Center, Massachusetts General Hospital, Harvard Medical School, Charlestown, United States
| | - Shyamala Maheswaran
- Cancer Center, Massachusetts General Hospital, Harvard Medical School, Charlestown, United States
| | - Daniel A Haber
- Cancer Center, Massachusetts General Hospital, Harvard Medical School, Charlestown, United States
| |
Collapse
|
157
|
Ping Y, Deng Y, Wang L, Zhang H, Zhang Y, Xu C, Zhao H, Fan H, Yu F, Xiao Y, Li X. Identifying core gene modules in glioblastoma based on multilayer factor-mediated dysfunctional regulatory networks through integrating multi-dimensional genomic data. Nucleic Acids Res 2015; 43:1997-2007. [PMID: 25653168 PMCID: PMC4344511 DOI: 10.1093/nar/gkv074] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
The driver genetic aberrations collectively regulate core cellular processes underlying cancer development. However, identifying the modules of driver genetic alterations and characterizing their functional mechanisms are still major challenges for cancer studies. Here, we developed an integrative multi-omics method CMDD to identify the driver modules and their affecting dysregulated genes through characterizing genetic alteration-induced dysregulated networks. Applied to glioblastoma (GBM), the CMDD identified a core gene module of 17 genes, including seven known GBM drivers, and their dysregulated genes. The module showed significant association with shorter survival of GBM. When classifying driver genes in the module into two gene sets according to their genetic alteration patterns, we found that one gene set directly participated in the glioma pathway, while the other indirectly regulated the glioma pathway, mostly, via their dysregulated genes. Both of the two gene sets were significant contributors to survival and helpful for classifying GBM subtypes, suggesting their critical roles in GBM pathogenesis. Also, by applying the CMDD to other six cancers, we identified some novel core modules associated with overall survival of patients. Together, these results demonstrate integrative multi-omics data can identify driver modules and uncover their dysregulated genes, which is useful for interpreting cancer genome.
Collapse
Affiliation(s)
- Yanyan Ping
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang 150086, China
| | - Yulan Deng
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang 150086, China
| | - Li Wang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang 150086, China
| | - Hongyi Zhang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang 150086, China
| | - Yong Zhang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang 150086, China
| | - Chaohan Xu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang 150086, China
| | - Hongying Zhao
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang 150086, China
| | - Huihui Fan
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang 150086, China
| | - Fulong Yu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang 150086, China
| | - Yun Xiao
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang 150086, China
| | - Xia Li
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang 150086, China
| |
Collapse
|
158
|
Repunte-Canonigo V, Shin W, Vendruscolo LF, Lefebvre C, van der Stap L, Kawamura T, Schlosburg JE, Alvarez M, Koob GF, Califano A, Sanna PP. Identifying candidate drivers of alcohol dependence-induced excessive drinking by assembly and interrogation of brain-specific regulatory networks. Genome Biol 2015; 16:68. [PMID: 25886852 PMCID: PMC4410476 DOI: 10.1186/s13059-015-0593-5] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2014] [Accepted: 01/21/2015] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND A systems biology approach based on the assembly and interrogation of gene regulatory networks, or interactomes, was used to study neuroadaptation processes associated with the transition to alcohol dependence at the molecular level. RESULTS Using a rat model of dependent and non-dependent alcohol self-administration, we reverse engineered a global transcriptional regulatory network during protracted abstinence, a period when relapse rates are highest. We then interrogated the network to identify master regulator genes that mechanistically regulate brain region-specific signatures associated with dependent and non-dependent alcohol self-administration. Among these, the gene coding for the glucocorticoid receptor was independently identified as a master regulator in multiple brain regions, including the medial prefrontal cortex, nucleus accumbens, central nucleus of the amygdala, and ventral tegmental area, consistent with the view that brain reward and stress systems are dysregulated during protracted abstinence. Administration of the glucocorticoid antagonist mifepristone in either the nucleus accumbens or ventral tegmental area selectively decreased dependent, excessive, alcohol self-administration in rats but had no effect on non-dependent, moderate, alcohol self-administration. CONCLUSIONS Our study suggests that assembly and analysis of regulatory networks is an effective strategy for the identification of key regulators of long-term neuroplastic changes within specific brain regions that play a functional role in alcohol dependence. More specifically, our results support a key role for regulatory networks downstream of the glucocorticoid receptor in excessive alcohol drinking during protracted alcohol abstinence.
Collapse
Affiliation(s)
- Vez Repunte-Canonigo
- Molecular and Integrative Neuroscience Department, The Scripps Research Institute, La Jolla, CA, USA.
| | - William Shin
- Department of Biological Sciences, Columbia University, New York, NY, 10027, USA. .,Department of Systems Biology, Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY, 10032, USA.
| | - Leandro F Vendruscolo
- Committee for the Neurobiology of Addictive Disorders, The Scripps Research Institute, La Jolla, CA, USA. .,Current affiliation: Intramural Research Program, NIDA-NIH, Baltimore, MD, 21224, USA.
| | - Celine Lefebvre
- Department of Systems Biology, Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY, 10032, USA. .,Current affiliation: Inserm Unit U981, Gustave Roussy Institute, Villejuif, France.
| | - Lena van der Stap
- Molecular and Integrative Neuroscience Department, The Scripps Research Institute, La Jolla, CA, USA.
| | - Tomoya Kawamura
- Molecular and Integrative Neuroscience Department, The Scripps Research Institute, La Jolla, CA, USA.
| | - Joel E Schlosburg
- Committee for the Neurobiology of Addictive Disorders, The Scripps Research Institute, La Jolla, CA, USA.
| | - Mariano Alvarez
- Department of Systems Biology, Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY, 10032, USA.
| | - George F Koob
- Committee for the Neurobiology of Addictive Disorders, The Scripps Research Institute, La Jolla, CA, USA. .,Current affiliation: National Institute on Alcohol Abuse and Alcoholism, Rockville, MD, 20852, USA.
| | - Andrea Califano
- Department of Systems Biology, Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY, 10032, USA. .,Department of Biomedical Informatics, Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY, 10032, USA. .,Institute for Cancer Genetics, Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY, 10032, USA. .,Department of Biochemistry and Molecular Biophysics, Hammer Health Sciences Center, Columbia University, New York, NY, 10032, USA. .,Cancer Regulatory Network Program, Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY, 10032, USA. .,The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA, 92037, USA.
| | - Pietro Paolo Sanna
- Molecular and Integrative Neuroscience Department, The Scripps Research Institute, La Jolla, CA, USA.
| |
Collapse
|
159
|
Xing Y, Wang H, Liu X, Yu X, Chen R, Wang C, Yu X, Sun L. Exploring the genes associated with the response to intravenous immunoglobulin in patients with Kawasaki disease using DNA microarray analysis. Exp Mol Pathol 2015; 98:7-12. [DOI: 10.1016/j.yexmp.2014.11.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2014] [Revised: 09/25/2014] [Accepted: 11/12/2014] [Indexed: 02/07/2023]
|
160
|
Dai Z, Guo D, Dai X, Xiong Y. Genome-wide analysis of transcription factor binding sites and their characteristic DNA structures. BMC Genomics 2015; 16 Suppl 3:S8. [PMID: 25708259 PMCID: PMC4331811 DOI: 10.1186/1471-2164-16-s3-s8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Background Transcription factors (TF) regulate gene expression by binding DNA regulatory regions. Transcription factor binding sites (TFBSs) are conserved not only in primary DNA sequences but also in DNA structures. However, the global relationship between TFs and their preferred DNA structures remains to be elucidated. Results In this paper, we have developed a computational method to generate a genome-wide landscape of TFs and their characteristic binding DNA structures in Saccharomyces cerevisiae. We revealed DNA structural features for different TFs. The structural conservation shows positional preference in TFBSs. Structural levels of DNA sequences are correlated with TF-DNA binding affinities. Conclusions We provided the genome-wide correspondences of TFs to DNA structures. Our findings will have implications in understanding TF regulatory mechanisms.
Collapse
|
161
|
Petolino JF. Genome editing in plants via designed zinc finger nucleases. IN VITRO CELLULAR & DEVELOPMENTAL BIOLOGY. PLANT : JOURNAL OF THE TISSUE CULTURE ASSOCIATION 2015; 51:1-8. [PMID: 25774080 PMCID: PMC4352198 DOI: 10.1007/s11627-015-9663-3] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/24/2014] [Accepted: 01/07/2015] [Indexed: 05/19/2023]
Abstract
The ability to create DNA double-strand breaks (DSBs) at specified genomic locations, which then stimulate the cell's naturally occurring DNA repair processes, has introduced intriguing possibilities for genetic modification. Zinc finger nucleases (ZFNs) are designed restriction enzymes consisting of a nonspecific cleavage domain fused to sequence-specific DNA binding domains. ZFN-mediated DSB formation at endogenous genomic loci followed by error-prone non-homologous end joining (NHEJ) repair can result in gene-specific mutations via nucleotide base pair insertions or deletions. Similarly, specific DNA sequence modifications can be made by providing donor DNA templates homologous to sequences flanking the cleavage site via homology-directed repair (HDR). Targeted deletions of intervening DNA sequence can be obtained by ZFNs used to create concurrent DSBs. Site-specific transgene integration into ZFN-induced DSBs is possible via either NHEJ or HDR. Genome editing can be used to enhance our basic understanding of plant gene function as well as modify and improve crop plants. As with conventional plant transformation technology, the efficiency of genome editing is absolutely dependent on the ability to initiate, maintain, and regenerate plant cell and tissue cultures.
Collapse
|
162
|
Yella VR, Bansal M. In silico Identification of Eukaryotic Promoters. SYSTEMS AND SYNTHETIC BIOLOGY 2015. [DOI: 10.1007/978-94-017-9514-2_4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
163
|
Ramsey SA, Vengrenyuk Y, Menon P, Podolsky I, Feig JE, Aderem A, Fisher EA, Gold ES. Epigenome-guided analysis of the transcriptome of plaque macrophages during atherosclerosis regression reveals activation of the Wnt signaling pathway. PLoS Genet 2014; 10:e1004828. [PMID: 25474352 PMCID: PMC4256277 DOI: 10.1371/journal.pgen.1004828] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2014] [Accepted: 10/15/2014] [Indexed: 11/19/2022] Open
Abstract
We report the first systems biology investigation of regulators controlling arterial plaque macrophage transcriptional changes in response to lipid lowering in vivo in two distinct mouse models of atherosclerosis regression. Transcriptome measurements from plaque macrophages from the Reversa mouse were integrated with measurements from an aortic transplant-based mouse model of plaque regression. Functional relevance of the genes detected as differentially expressed in plaque macrophages in response to lipid lowering in vivo was assessed through analysis of gene functional annotations, overlap with in vitro foam cell studies, and overlap of associated eQTLs with human atherosclerosis/CAD risk SNPs. To identify transcription factors that control plaque macrophage responses to lipid lowering in vivo, we used an integrative strategy – leveraging macrophage epigenomic measurements – to detect enrichment of transcription factor binding sites upstream of genes that are differentially expressed in plaque macrophages during regression. The integrated analysis uncovered eight transcription factor binding site elements that were statistically overrepresented within the 5′ regulatory regions of genes that were upregulated in plaque macrophages in the Reversa model under maximal regression conditions and within the 5′ regulatory regions of genes that were upregulated in the aortic transplant model during regression. Of these, the TCF/LEF binding site was present in promoters of upregulated genes related to cell motility, suggesting that the canonical Wnt signaling pathway may be activated in plaque macrophages during regression. We validated this network-based prediction by demonstrating that β-catenin expression is higher in regressing (vs. control group) plaques in both regression models, and we further demonstrated that stimulation of canonical Wnt signaling increases macrophage migration in vitro. These results suggest involvement of canonical Wnt signaling in macrophage emigration from the plaque during lipid lowering-induced regression, and they illustrate the discovery potential of an epigenome-guided, systems approach to understanding atherosclerosis regression. Atherosclerosis, a progressive accumulation of lipid-rich plaque within arteries, is an inflammatory disease in which the response of macrophages (a key cell type of the innate immune system) to plasma lipoproteins plays a central role. In humans, the goal of significantly reducing already-established plaque through drug treatments, including statins, remains elusive. In mice, atherosclerosis can be reversed by experimental manipulations that lower circulating lipid levels. A common feature of many regression models is that macrophages transition to a less inflammatory state and emigrate from the plaque. While the molecular regulators that control these responses are largely unknown, we hypothesized that by integrating global measurements of macrophage gene expression in regressing plaques with measurements of the macrophage chromatin landscape, we could identify key molecules that control macrophage responses to the lowering of circulating lipid levels. Our systems biology analysis of plaque macrophages yielded a network in which the Wnt signaling pathway emerged as a candidate upstream regulator. Wnt signaling is known to affect both inflammation and the ability of macrophages to migrate from one location to another, and our targeted validation studies provide evidence that Wnt signaling is increased in plaque macrophages during regression. Our findings both demonstrate the power of a systems approach to uncover candidate regulators of regression and to identify a potential new therapeutic target.
Collapse
MESH Headings
- Animals
- Cells, Cultured
- Epigenesis, Genetic/drug effects
- Epigenesis, Genetic/physiology
- Female
- Gene Expression Profiling
- Genome/drug effects
- Hypolipidemic Agents/pharmacology
- Hypolipidemic Agents/therapeutic use
- Macrophages/drug effects
- Macrophages/metabolism
- Macrophages/pathology
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Microarray Analysis
- Plaque, Atherosclerotic/drug therapy
- Plaque, Atherosclerotic/genetics
- Plaque, Atherosclerotic/metabolism
- Plaque, Atherosclerotic/pathology
- Receptors, LDL/genetics
- Remission Induction
- Transcriptome/drug effects
- Wnt Signaling Pathway/drug effects
- Wnt Signaling Pathway/genetics
Collapse
Affiliation(s)
- Stephen A. Ramsey
- Department of Biomedical Sciences and School of Electrical Engineering and Computer Science, Oregon State University, Corvallis, Oregon, United States of America
| | - Yuliya Vengrenyuk
- Division of Cardiology, School of Medicine, New York University, New York, New York, United States of America
| | - Prashanthi Menon
- Division of Cardiology, School of Medicine, New York University, New York, New York, United States of America
| | - Irina Podolsky
- Seattle Biomedical Research Institute, Seattle, Washington, United States of America
| | - Jonathan E. Feig
- Division of Cardiology, School of Medicine, New York University, New York, New York, United States of America
| | - Alan Aderem
- Seattle Biomedical Research Institute, Seattle, Washington, United States of America
| | - Edward A. Fisher
- Division of Cardiology, School of Medicine, New York University, New York, New York, United States of America
- * E-mail: (EAF); (ESG)
| | - Elizabeth S. Gold
- Seattle Biomedical Research Institute, Seattle, Washington, United States of America
- * E-mail: (EAF); (ESG)
| |
Collapse
|
164
|
Castro-García FP, Corral-Jara KF, Escobedo-Melendez G, Sandoval-Hernandez MA, Rosenstein Y, Roman S, Panduro A, Fierro NA. Conjugated bilirubin affects cytokine profiles in hepatitis A virus infection by modulating function of signal transducer and activator of transcription factors. Immunology 2014; 143:578-587. [PMID: 24943111 PMCID: PMC4253506 DOI: 10.1111/imm.12336] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2014] [Revised: 06/11/2014] [Accepted: 06/13/2014] [Indexed: 12/24/2022] Open
Abstract
Hepatitis A virus (HAV) infection is the major cause of acute liver failure in paediatric patients. The clinical spectrum of infection is variable, and liver injury is determined by altered hepatic enzyme function and bilirubin concentration. We recently reported differences in cytokine profiles between distinct HAV-induced clinical courses, and bilirubin has been recognized as a potential immune-modulator. However, how bilirubin may affect cytokine profiles underlying the variability in the course of infection has not been determined. Herein, we used a transcription factor (TF) binding site identification approach to retrospectively analyse cytokine expression in HAV-infected children and to predict the entire set of TFs associated with the expression of specific cytokine profiles. The results suggested that modulation of the activity of signal transducers and activators of transcription proteins (STATs) may play a central role during HAV infection. This led us to compare the degree of STAT phosphorylation in peripheral blood lymphoid cells (PBLCs) from paediatric patients with distinct levels of conjugated bilirubin (CB). Low CB levels in sera were associated with increased STAT-1 and STAT-5 phosphorylation. A positive correlation was observed between the serum interleukin-6 (IL-6) content and CB values, whereas higher levels of CB correlated with reduced serum IL-8 values and with a reduction in the proportion of PBLCs positive for STAT-5 phosphorylation. When CB was used to stimulate patients' PBLCs in vitro, the levels of IL-6 and tumour necrosis factor-α were increased. The data showed that bilirubin plays a role in STAT function and affects cytokine profile expression during HAV infection.
Collapse
Affiliation(s)
- Flor P Castro-García
- Unidad de Inmunovirología, Hospital Civil de Guadalajara Fray Antonio AlcaldeGuadalajara, Mexico
- Centro Universitario de Ciencias de la Salud, Universidad de GuadalajaraGuadalajara, Mexico
| | - Karla F Corral-Jara
- Unidad de Inmunovirología, Hospital Civil de Guadalajara Fray Antonio AlcaldeGuadalajara, Mexico
- Centro Universitario de Ciencias de la Salud, Universidad de GuadalajaraGuadalajara, Mexico
| | | | - Monserrat A Sandoval-Hernandez
- Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de MéxicoCuernavaca, Mexico
| | - Yvonne Rosenstein
- Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de MéxicoCuernavaca, Mexico
| | - Sonia Roman
- Centro Universitario de Ciencias de la Salud, Universidad de GuadalajaraGuadalajara, Mexico
- Servicio de Biología Molecular, Hospital Civil de Guadalajara Fray Antonio AlcaldeGuadalajara, Mexico
| | - Arturo Panduro
- Centro Universitario de Ciencias de la Salud, Universidad de GuadalajaraGuadalajara, Mexico
- Servicio de Biología Molecular, Hospital Civil de Guadalajara Fray Antonio AlcaldeGuadalajara, Mexico
| | - Nora A Fierro
- Unidad de Inmunovirología, Hospital Civil de Guadalajara Fray Antonio AlcaldeGuadalajara, Mexico
- Centro Universitario de Ciencias de la Salud, Universidad de GuadalajaraGuadalajara, Mexico
- Servicio de Biología Molecular, Hospital Civil de Guadalajara Fray Antonio AlcaldeGuadalajara, Mexico
| |
Collapse
|
165
|
Yang WS, Chadalapaka G, Cho SG, Lee SO, Jin UH, Jutooru I, Choi K, Leung YK, Ho SM, Safe S, Kim K. The transcriptional repressor ZBTB4 regulates EZH2 through a MicroRNA-ZBTB4-specificity protein signaling axis. Neoplasia 2014; 16:1059-1069. [PMID: 25499219 PMCID: PMC4309261 DOI: 10.1016/j.neo.2014.09.011] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2014] [Revised: 09/22/2014] [Accepted: 09/29/2014] [Indexed: 10/28/2022]
Abstract
ZBTB4 is a transcriptional repressor and examination of publically-available microarray data sets demonstrated an inverse relationship in the prognostic value and expression of ZBTB4 and the histone methyltransferase EZH2 in tumors from breast cancer patients. The possibility of functional interactions between EZH2 and ZBTB4 was investigated in breast cancer cells and the results showed that EZH2 is directly suppressed by ZBTB4 which in turn is regulated (suppressed) by miR-106b and other paralogues from the miR-17-92, miR-106b-25 and miR-106a-363 clusters that are highly expressed in breast and other tumors. ZBTB4 also acts a suppressor of specificity protein (Sp) transcription factors Sp1, Sp3 and Sp4, and RNA interference studies show that Sp proteins are required for EZH2 expression. The prediction analysis results from breast cancer patient array data sets confirm an association of Sp1-dependent EZH2 gene signature with decreased survival of breast cancer patients. Disruption of oncogenic miR-ZBTB4 signaling axis by anticancer agent such as betulinic acid that induce down-regulation of Sp proteins in breast cancer cells resulted in inhibition of tumor growth and colonization of breast cancer cells in a mouse model. Thus, EZH2 is reciprocally regulated by a novel signaling network consisting of Sp proteins, oncogenic miRs and ZBTB4, and modulation of this gene network is a novel therapeutic approach for treatment of breast cancer and possibly other cancers.
Collapse
Affiliation(s)
- Won Seok Yang
- Department of Environmental Health, University of Cincinnati, College of Medicine, 3223 Eden Ave., Cincinnati, OH 45267
| | - Gayathri Chadalapaka
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, 4466 Texas A&M University, College Station, TX 77843
| | - Sung-Gook Cho
- Department of Preventive Medicine, College of Korean Medicine, Kyung Hee University, 1 Hoegi, Dongdaemun-gu, Seoul 130-701, Republic of Korea
| | - Syng-Ook Lee
- Institute of Biosciences and Technology, Texas A&M Health Science Center, 2121 W. Holcombe Blvd., Houston, TX 77030
| | - Un-Ho Jin
- Institute of Biosciences and Technology, Texas A&M Health Science Center, 2121 W. Holcombe Blvd., Houston, TX 77030
| | - Indira Jutooru
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, 4466 Texas A&M University, College Station, TX 77843
| | - Kwangmin Choi
- Division of Experimental Biology and Cancer Biology, Cincinnati Children's Hospital medical Center, Cincinnati, OH 45229
| | - Yuet-Kin Leung
- Department of Environmental Health, University of Cincinnati, College of Medicine, 3223 Eden Ave., Cincinnati, OH 45267
| | - Shuk-Mei Ho
- Department of Environmental Health, University of Cincinnati, College of Medicine, 3223 Eden Ave., Cincinnati, OH 45267
| | - Stephen Safe
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, 4466 Texas A&M University, College Station, TX 77843; Institute of Biosciences and Technology, Texas A&M Health Science Center, 2121 W. Holcombe Blvd., Houston, TX 77030
| | - Kyounghyun Kim
- Department of Environmental Health, University of Cincinnati, College of Medicine, 3223 Eden Ave., Cincinnati, OH 45267
| |
Collapse
|
166
|
Azghandi S, Prell C, van der Laan SW, Schneider M, Malik R, Berer K, Gerdes N, Pasterkamp G, Weber C, Haffner C, Dichgans M. Deficiency of the stroke relevant HDAC9 gene attenuates atherosclerosis in accord with allele-specific effects at 7p21.1. Stroke 2014; 46:197-202. [PMID: 25388417 DOI: 10.1161/strokeaha.114.007213] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
BACKGROUND AND PURPOSE Recent genome-wide association studies identified the histone deacetylase 9 (HDAC9) gene region as a major risk locus for large-vessel stroke and coronary artery disease. However, the mechanisms linking variants at this locus to vascular risk are poorly understood. In this study, we investigated the candidacy and directionality of HDAC9 in atherosclerosis and analyzed associations between risk alleles at 7p21.1 and plaque characteristics. METHODS Allele-dependent expression of HDAC9 was analyzed in human peripheral blood mononuclear cells of healthy donors. Effects of HDAC9 deficiency on atherosclerotic plaques were investigated in 18- and 28-week-old ApoE(-/-) mice by histology and immunohistochemistry. We further performed detailed plaque phenotyping and genotyping of rs2107595, the lead single-nucleotide polymorphism for large-vessel stroke, in carotid endarterectomy samples of 1858 subjects from the Athero-Express study. RESULTS Gene expression studies in peripheral blood mononuclear cells revealed increased mRNA levels of HDAC9 but not of neighboring genes (TWIST1/FERD3L) in risk allele carriers of rs2107595. Compared with HDAC9(+/+)ApoE(-/-) mice, HDAC9(-/-)ApoE(-/-) mice exhibited markedly reduced lesion sizes throughout atherosclerotic aortas and significantly less advanced lesions. The proportion of Mac3-positive macrophages was higher in plaques from HDAC9(-/-)ApoE(-/-) mice, but this was largely because of a lower proportion of advanced lesions. Analysis of human atherosclerotic plaques revealed no association between rs2107595 and specific plaque characteristics. CONCLUSIONS Our results suggest that HDAC9 represents the disease-relevant gene at the stroke and coronary artery disease risk locus on 7p21.1, and that risk alleles in this region mediate their effects through increased HDAC9 expression. Targeted inhibition of HDAC9 might be a viable strategy to prevent atherosclerosis.
Collapse
Affiliation(s)
- Sepiede Azghandi
- From the Institute for Stroke and Dementia Research, Klinikum der Universität München, Munich, Germany (S.A., C.P., M.S., R.M., C.H., M.D.); Divisions Heart and Lungs and Laboratories and Pharmacy, University Medical Center Utrecht, Utrecht, The Netherlands (S.W.v.d.L., G.P.); Department of Neuroimmunology, Max Planck Institute of Neurobiology, Martinsried, Germany (K.B.); Institute for Cardiovascular Prevention, Ludwig-Maximilians-University, Munich, Germany (N.G., C.W.); and Munich Cluster for Systems Neurology (SyNergy), Munich, Germany (M.D.)
| | - Caroline Prell
- From the Institute for Stroke and Dementia Research, Klinikum der Universität München, Munich, Germany (S.A., C.P., M.S., R.M., C.H., M.D.); Divisions Heart and Lungs and Laboratories and Pharmacy, University Medical Center Utrecht, Utrecht, The Netherlands (S.W.v.d.L., G.P.); Department of Neuroimmunology, Max Planck Institute of Neurobiology, Martinsried, Germany (K.B.); Institute for Cardiovascular Prevention, Ludwig-Maximilians-University, Munich, Germany (N.G., C.W.); and Munich Cluster for Systems Neurology (SyNergy), Munich, Germany (M.D.)
| | - Sander W van der Laan
- From the Institute for Stroke and Dementia Research, Klinikum der Universität München, Munich, Germany (S.A., C.P., M.S., R.M., C.H., M.D.); Divisions Heart and Lungs and Laboratories and Pharmacy, University Medical Center Utrecht, Utrecht, The Netherlands (S.W.v.d.L., G.P.); Department of Neuroimmunology, Max Planck Institute of Neurobiology, Martinsried, Germany (K.B.); Institute for Cardiovascular Prevention, Ludwig-Maximilians-University, Munich, Germany (N.G., C.W.); and Munich Cluster for Systems Neurology (SyNergy), Munich, Germany (M.D.)
| | - Manuela Schneider
- From the Institute for Stroke and Dementia Research, Klinikum der Universität München, Munich, Germany (S.A., C.P., M.S., R.M., C.H., M.D.); Divisions Heart and Lungs and Laboratories and Pharmacy, University Medical Center Utrecht, Utrecht, The Netherlands (S.W.v.d.L., G.P.); Department of Neuroimmunology, Max Planck Institute of Neurobiology, Martinsried, Germany (K.B.); Institute for Cardiovascular Prevention, Ludwig-Maximilians-University, Munich, Germany (N.G., C.W.); and Munich Cluster for Systems Neurology (SyNergy), Munich, Germany (M.D.)
| | - Rainer Malik
- From the Institute for Stroke and Dementia Research, Klinikum der Universität München, Munich, Germany (S.A., C.P., M.S., R.M., C.H., M.D.); Divisions Heart and Lungs and Laboratories and Pharmacy, University Medical Center Utrecht, Utrecht, The Netherlands (S.W.v.d.L., G.P.); Department of Neuroimmunology, Max Planck Institute of Neurobiology, Martinsried, Germany (K.B.); Institute for Cardiovascular Prevention, Ludwig-Maximilians-University, Munich, Germany (N.G., C.W.); and Munich Cluster for Systems Neurology (SyNergy), Munich, Germany (M.D.)
| | - Kerstin Berer
- From the Institute for Stroke and Dementia Research, Klinikum der Universität München, Munich, Germany (S.A., C.P., M.S., R.M., C.H., M.D.); Divisions Heart and Lungs and Laboratories and Pharmacy, University Medical Center Utrecht, Utrecht, The Netherlands (S.W.v.d.L., G.P.); Department of Neuroimmunology, Max Planck Institute of Neurobiology, Martinsried, Germany (K.B.); Institute for Cardiovascular Prevention, Ludwig-Maximilians-University, Munich, Germany (N.G., C.W.); and Munich Cluster for Systems Neurology (SyNergy), Munich, Germany (M.D.)
| | - Norbert Gerdes
- From the Institute for Stroke and Dementia Research, Klinikum der Universität München, Munich, Germany (S.A., C.P., M.S., R.M., C.H., M.D.); Divisions Heart and Lungs and Laboratories and Pharmacy, University Medical Center Utrecht, Utrecht, The Netherlands (S.W.v.d.L., G.P.); Department of Neuroimmunology, Max Planck Institute of Neurobiology, Martinsried, Germany (K.B.); Institute for Cardiovascular Prevention, Ludwig-Maximilians-University, Munich, Germany (N.G., C.W.); and Munich Cluster for Systems Neurology (SyNergy), Munich, Germany (M.D.)
| | - Gerard Pasterkamp
- From the Institute for Stroke and Dementia Research, Klinikum der Universität München, Munich, Germany (S.A., C.P., M.S., R.M., C.H., M.D.); Divisions Heart and Lungs and Laboratories and Pharmacy, University Medical Center Utrecht, Utrecht, The Netherlands (S.W.v.d.L., G.P.); Department of Neuroimmunology, Max Planck Institute of Neurobiology, Martinsried, Germany (K.B.); Institute for Cardiovascular Prevention, Ludwig-Maximilians-University, Munich, Germany (N.G., C.W.); and Munich Cluster for Systems Neurology (SyNergy), Munich, Germany (M.D.)
| | - Christian Weber
- From the Institute for Stroke and Dementia Research, Klinikum der Universität München, Munich, Germany (S.A., C.P., M.S., R.M., C.H., M.D.); Divisions Heart and Lungs and Laboratories and Pharmacy, University Medical Center Utrecht, Utrecht, The Netherlands (S.W.v.d.L., G.P.); Department of Neuroimmunology, Max Planck Institute of Neurobiology, Martinsried, Germany (K.B.); Institute for Cardiovascular Prevention, Ludwig-Maximilians-University, Munich, Germany (N.G., C.W.); and Munich Cluster for Systems Neurology (SyNergy), Munich, Germany (M.D.)
| | - Christof Haffner
- From the Institute for Stroke and Dementia Research, Klinikum der Universität München, Munich, Germany (S.A., C.P., M.S., R.M., C.H., M.D.); Divisions Heart and Lungs and Laboratories and Pharmacy, University Medical Center Utrecht, Utrecht, The Netherlands (S.W.v.d.L., G.P.); Department of Neuroimmunology, Max Planck Institute of Neurobiology, Martinsried, Germany (K.B.); Institute for Cardiovascular Prevention, Ludwig-Maximilians-University, Munich, Germany (N.G., C.W.); and Munich Cluster for Systems Neurology (SyNergy), Munich, Germany (M.D.)
| | - Martin Dichgans
- From the Institute for Stroke and Dementia Research, Klinikum der Universität München, Munich, Germany (S.A., C.P., M.S., R.M., C.H., M.D.); Divisions Heart and Lungs and Laboratories and Pharmacy, University Medical Center Utrecht, Utrecht, The Netherlands (S.W.v.d.L., G.P.); Department of Neuroimmunology, Max Planck Institute of Neurobiology, Martinsried, Germany (K.B.); Institute for Cardiovascular Prevention, Ludwig-Maximilians-University, Munich, Germany (N.G., C.W.); and Munich Cluster for Systems Neurology (SyNergy), Munich, Germany (M.D.).
| |
Collapse
|
167
|
Regression analysis of combined gene expression regulation in acute myeloid leukemia. PLoS Comput Biol 2014; 10:e1003908. [PMID: 25340776 PMCID: PMC4207489 DOI: 10.1371/journal.pcbi.1003908] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2014] [Accepted: 09/13/2014] [Indexed: 12/04/2022] Open
Abstract
Gene expression is a combinatorial function of genetic/epigenetic factors such as copy number variation (CNV), DNA methylation (DM), transcription factors (TF) occupancy, and microRNA (miRNA) post-transcriptional regulation. At the maturity of microarray/sequencing technologies, large amounts of data measuring the genome-wide signals of those factors became available from Encyclopedia of DNA Elements (ENCODE) and The Cancer Genome Atlas (TCGA). However, there is a lack of an integrative model to take full advantage of these rich yet heterogeneous data. To this end, we developed RACER (Regression Analysis of Combined Expression Regulation), which fits the mRNA expression as response using as explanatory variables, the TF data from ENCODE, and CNV, DM, miRNA expression signals from TCGA. Briefly, RACER first infers the sample-specific regulatory activities by TFs and miRNAs, which are then used as inputs to infer specific TF/miRNA-gene interactions. Such a two-stage regression framework circumvents a common difficulty in integrating ENCODE data measured in generic cell-line with the sample-specific TCGA measurements. As a case study, we integrated Acute Myeloid Leukemia (AML) data from TCGA and the related TF binding data measured in K562 from ENCODE. As a proof-of-concept, we first verified our model formalism by 10-fold cross-validation on predicting gene expression. We next evaluated RACER on recovering known regulatory interactions, and demonstrated its superior statistical power over existing methods in detecting known miRNA/TF targets. Additionally, we developed a feature selection procedure, which identified 18 regulators, whose activities clustered consistently with cytogenetic risk groups. One of the selected regulators is miR-548p, whose inferred targets were significantly enriched for leukemia-related pathway, implicating its novel role in AML pathogenesis. Moreover, survival analysis using the inferred activities identified C-Fos as a potential AML prognostic marker. Together, we provided a novel framework that successfully integrated the TCGA and ENCODE data in revealing AML-specific regulatory program at global level. Recent studies from The Cancer Genome Atlas (TCGA) showed that most Acute Myeloid Leukemia (AML) patients lack DNA mutations, which can potentially explain the tumorigenesis, and motivated a systematic approach to elucidate aberrant molecular signatures at the transcriptional and epigenetic levels. Using recently available data from two large consortia namely Encyclopedia of DNA Elements and TCGA, we developed a novel computational model to infer the regulatory activities of the expression regulators and their target genes in AML samples. Our analysis revealed 18 regulators whose dysregulation contributed significantly to explaining the global mRNA expression changes. Encouragingly, the inferred activities of these regulatory features followed a consistent pattern with cytogenetic phenotypes of the AML patients. Among these regulators, we identified microRNA hsa-miR-548p, whose regulatory relationships with leukemia-related genes including YY1 suggest its novel role in AML pathogenesis. Additionally, we discovered that the inferred activities of transcription factor C-Fos can be used as a prognostic marker to characterize survival rate of the AML patients. Together, we demonstrated an effective model that can integrate useful information from a large amount of heterogeneous data to dissect regulatory effects. Furthermore, the novel biological findings from this study may be constructive to future experimental research in AML.
Collapse
|
168
|
Liu J, Wang X, Li J, Wang H, Wei G, Yan J. Reconstruction of the gene regulatory network involved in the sonic hedgehog pathway with a potential role in early development of the mouse brain. PLoS Comput Biol 2014; 10:e1003884. [PMID: 25299227 PMCID: PMC4191885 DOI: 10.1371/journal.pcbi.1003884] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2014] [Accepted: 08/28/2014] [Indexed: 11/18/2022] Open
Abstract
The Sonic hedgehog (Shh) signaling pathway is crucial for pattern formation in early central nervous system development. By systematically analyzing high-throughput in situ hybridization data of E11.5 mouse brain, we found that Shh and its receptor Ptch1 define two adjacent mutually exclusive gene expression domains: Shh+Ptch1− and Shh−Ptch1+. These two domains are associated respectively with Foxa2 and Gata3, two transcription factors that play key roles in specifying them. Gata3 ChIP-seq experiments and RNA-seq assays on Gata3-knockdown cells revealed that Gata3 up-regulates the genes that are enriched in the Shh−Ptch1+ domain. Important Gata3 targets include Slit2 and Slit3, which are involved in the process of axon guidance, as well as Slc18a1, Th and Qdpr, which are associated with neurotransmitter synthesis and release. By contrast, Foxa2 both up-regulates the genes expressed in the Shh+Ptch1− domain and down-regulates the genes characteristic of the Shh−Ptch1+ domain. From these and other data, we were able to reconstruct a gene regulatory network governing both domains. Our work provides the first genome-wide characterization of the gene regulatory network involved in the Shh pathway that underlies pattern formation in the early mouse brain. Recent large-scale projects of high-throughput in situ hybridization (ISH) have generated a wealth of spatiotemporal information on gene expression patterns in the early mouse brain. We have developed a computational approach that combines publicly available high-throughput ISH data with our own experimental data to investigate gene regulation, involving signal molecules and transcription factors (TFs), during early brain development. The analysis indicates that two key TFs, Foxa2 and Gata3, play antagonizing roles in the formation of two mutually exclusive domains established by the Sonic hedgehog signaling pathway in the developing mouse brain. Further ChIP-seq and RNA-seq experiments support this hypothesis and have identified novel target genes of Gata3, including the axon guidance regulators Slit2 and Slit3 as well as three neurotransmitter-associated genes, Slc18a1, Th and Qdpr. The findings have allowed us to reconstruct the gene regulatory network brought into play by the Sonic hedgehog pathway that mediates early mouse brain development.
Collapse
Affiliation(s)
- Jinhua Liu
- CAS-MPG Partner Institute for Computational Biology, Shanghai Institutes of Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Xuelong Wang
- CAS-MPG Partner Institute for Computational Biology, Shanghai Institutes of Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Juan Li
- CAS-MPG Partner Institute for Computational Biology, Shanghai Institutes of Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Haifang Wang
- CAS-MPG Partner Institute for Computational Biology, Shanghai Institutes of Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Gang Wei
- CAS-MPG Partner Institute for Computational Biology, Shanghai Institutes of Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Jun Yan
- CAS-MPG Partner Institute for Computational Biology, Shanghai Institutes of Biological Sciences, Chinese Academy of Sciences, Shanghai, China
- * E-mail:
| |
Collapse
|
169
|
Navarro C, Lopez FJ, Cano C, Garcia-Alcalde F, Blanco A. CisMiner: genome-wide in-silico cis-regulatory module prediction by fuzzy itemset mining. PLoS One 2014; 9:e108065. [PMID: 25268582 PMCID: PMC4182448 DOI: 10.1371/journal.pone.0108065] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2014] [Accepted: 08/25/2014] [Indexed: 01/18/2023] Open
Abstract
Eukaryotic gene control regions are known to be spread throughout non-coding DNA sequences which may appear distant from the gene promoter. Transcription factors are proteins that coordinately bind to these regions at transcription factor binding sites to regulate gene expression. Several tools allow to detect significant co-occurrences of closely located binding sites (cis-regulatory modules, CRMs). However, these tools present at least one of the following limitations: 1) scope limited to promoter or conserved regions of the genome; 2) do not allow to identify combinations involving more than two motifs; 3) require prior information about target motifs. In this work we present CisMiner, a novel methodology to detect putative CRMs by means of a fuzzy itemset mining approach able to operate at genome-wide scale. CisMiner allows to perform a blind search of CRMs without any prior information about target CRMs nor limitation in the number of motifs. CisMiner tackles the combinatorial complexity of genome-wide cis-regulatory module extraction using a natural representation of motif combinations as itemsets and applying the Top-Down Fuzzy Frequent- Pattern Tree algorithm to identify significant itemsets. Fuzzy technology allows CisMiner to better handle the imprecision and noise inherent to regulatory processes. Results obtained for a set of well-known binding sites in the S. cerevisiae genome show that our method yields highly reliable predictions. Furthermore, CisMiner was also applied to putative in-silico predicted transcription factor binding sites to identify significant combinations in S. cerevisiae and D. melanogaster, proving that our approach can be further applied genome-wide to more complex genomes. CisMiner is freely accesible at: http://genome2.ugr.es/cisminer. CisMiner can be queried for the results presented in this work and can also perform a customized cis-regulatory module prediction on a query set of transcription factor binding sites provided by the user.
Collapse
Affiliation(s)
- Carmen Navarro
- Department of Computer Science and AI, University of Granada, Granada, Spain
| | - Francisco J. Lopez
- Andalusian Human Genome Sequencing Centre (CASEGH), Medical Genome Project (MGP), Sevilla, Spain
| | - Carlos Cano
- Department of Computer Science and AI, University of Granada, Granada, Spain
| | | | - Armando Blanco
- Department of Computer Science and AI, University of Granada, Granada, Spain
| |
Collapse
|
170
|
Reduced Krüppel-like factor 2 expression may aggravate the endothelial injury of diabetic nephropathy. Kidney Int 2014; 87:382-95. [PMID: 25185079 PMCID: PMC4312548 DOI: 10.1038/ki.2014.286] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2014] [Revised: 06/18/2014] [Accepted: 07/10/2014] [Indexed: 12/15/2022]
Abstract
Kruppel-like Factor 2 (KLF2), a shear-stress inducible transcription factor, has endoprotective effects. In streptozotocin-induced diabetic rats, we found that glomerular Klf2 expression was reduced in comparison to non-diabetic rats. However, normalization of hyperglycemia by insulin treatment increased Klf2 expression to a level higher than that of non-diabetic rats. Consistent with this, we found that Klf2 expression was suppressed by high glucose but increased by insulin in cultured endothelial cells. To determine the role of KLF2 in streptozotocin-induced diabetic nephropathy, we used endothelial cell-specific Klf2 heterozygous knockout mice and found that diabetic knockout mice developed more kidney/glomerular hypertrophy and proteinuria than diabetic wide type mice. Glomerular expression of Vegfa, Flk1, and angiopoietin 2 increased but expression of Flt1, Tie2, and angiopoietin 1 decreased in diabetic knockout compared to diabetic wide type mice. Glomerular expression of ZO-1, glycocalyx, and eNOS was also decreased in diabetic knockout compared to diabetic wide type mice. These data suggest knockdown of Klf2 expression in the endothelial cells induced more endothelial cell injury. Interestingly, podocyte injury was also more prominent in diabetic knockout compared to diabetic wide type mice, indicating a crosstalk between these two cell types. Thus, KLF2 may play a role in glomerular endothelial cell injury in early diabetic nephropathy.
Collapse
|
171
|
Wang C, Gong B, Bushel PR, Thierry-Mieg J, Thierry-Mieg D, Xu J, Fang H, Hong H, Shen J, Su Z, Meehan J, Li X, Yang L, Li H, Łabaj PP, Kreil DP, Megherbi D, Florian C, Gaj S, van Delft J, Kleinjans J, Scherer A, Viswanath D, Wang J, Yang Y, Qian HR, Lancashire LJ, Bessarabova M, Nikolsky Y, Furlanello C, Chierici M, Albanese D, Jurman G, Riccadonna S, Filosi M, Visintainer R, Zhang KK, Li J, Hsieh JH, Svoboda DL, Fuscoe JC, Deng Y, Shi L, Paules RS, Auerbach SS, Tong W. The concordance between RNA-seq and microarray data depends on chemical treatment and transcript abundance. Nat Biotechnol 2014; 32:926-32. [PMID: 25150839 PMCID: PMC4243706 DOI: 10.1038/nbt.3001] [Citation(s) in RCA: 368] [Impact Index Per Article: 33.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2013] [Accepted: 07/29/2014] [Indexed: 12/22/2022]
Abstract
The concordance of RNA-sequencing (RNA-seq) with microarrays for genome-wide analysis of differential gene expression has not been rigorously assessed using a range of chemical treatment conditions. Here we use a comprehensive study design to generate Illumina RNA-seq and Affymetrix microarray data from the same liver samples of rats exposed in triplicate to varying degrees of perturbation by 27 chemicals representing multiple modes of action (MOAs). The cross-platform concordance in terms of differentially expressed genes (DEGs) or enriched pathways is linearly correlated with treatment effect size (R(2)0.8). Furthermore, the concordance is also affected by transcript abundance and biological complexity of the MOA. RNA-seq outperforms microarray (93% versus 75%) in DEG verification as assessed by quantitative PCR, with the gain mainly due to its improved accuracy for low-abundance transcripts. Nonetheless, classifiers to predict MOAs perform similarly when developed using data from either platform. Therefore, the endpoint studied and its biological complexity, transcript abundance and the genomic application are important factors in transcriptomic research and for clinical and regulatory decision making.
Collapse
Affiliation(s)
- Charles Wang
- Center for Genomics and Division of Microbiology & Molecular Genetics, School of Medicine, Loma Linda University, Loma Linda, California, USA
| | - Binsheng Gong
- Division of Bioinformatics and Biostatistics, National Center for Toxicological Research, US Food and Drug Administration, Jefferson, Arkansas, USA
| | - Pierre R. Bushel
- Microarray and Genome Informatics Group, National Institute of Environmental Health Sciences, RTP, North Carolina, USA
- Biostatistics Branch, National Institute of Environmental Health Sciences, RTP, North Carolina, USA
| | - Jean Thierry-Mieg
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, Maryland, USA
| | - Danielle Thierry-Mieg
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, Maryland, USA
| | - Joshua Xu
- Division of Bioinformatics and Biostatistics, National Center for Toxicological Research, US Food and Drug Administration, Jefferson, Arkansas, USA
| | - Hong Fang
- The Office of Scientific Coordination, National Center for Toxicological Research, US Food and Drug Administration, Jefferson, Arkansas, USA
| | - Huixiao Hong
- Division of Bioinformatics and Biostatistics, National Center for Toxicological Research, US Food and Drug Administration, Jefferson, Arkansas, USA
| | - Jie Shen
- Division of Bioinformatics and Biostatistics, National Center for Toxicological Research, US Food and Drug Administration, Jefferson, Arkansas, USA
| | - Zhenqiang Su
- Division of Bioinformatics and Biostatistics, National Center for Toxicological Research, US Food and Drug Administration, Jefferson, Arkansas, USA
| | - Joe Meehan
- Division of Bioinformatics and Biostatistics, National Center for Toxicological Research, US Food and Drug Administration, Jefferson, Arkansas, USA
| | - Xiaojin Li
- Functional Genomics Core, Department of Molecular Medicine, Beckman Research Institute, City of Hope, Duarte, California, USA
| | - Lu Yang
- Functional Genomics Core, Department of Molecular Medicine, Beckman Research Institute, City of Hope, Duarte, California, USA
| | - Haiqing Li
- Functional Genomics Core, Department of Molecular Medicine, Beckman Research Institute, City of Hope, Duarte, California, USA
| | | | - David P. Kreil
- Boku University Vienna, Austria
- Life Sciences, University of Warwick, Coventry, U.K
| | - Dalila Megherbi
- CMINDS Research Center, Department of Electrical and Computer Engineering, Francis College of Engineering, University of Massachusetts, Lowell, Massachusetts, USA
| | - Caiment Florian
- Department of Toxicogenomics, Maastricht University, Maastricht, The Netherlands
| | - Stan Gaj
- Department of Toxicogenomics, Maastricht University, Maastricht, The Netherlands
| | - Joost van Delft
- Department of Toxicogenomics, Maastricht University, Maastricht, The Netherlands
| | - Jos Kleinjans
- Department of Toxicogenomics, Maastricht University, Maastricht, The Netherlands
| | - Andreas Scherer
- Australian Genome Research Facility Ltd, The Walter and Eliza Hall Institute of Medical Research, Melbourne, Australia
| | | | - Jian Wang
- Research Informatics and Statistics, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, Indiana, USA
| | - Yong Yang
- Research Informatics and Statistics, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, Indiana, USA
| | - Hui-Rong Qian
- Research Informatics and Statistics, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, Indiana, USA
| | | | | | - Yuri Nikolsky
- Thomson Reuters, IP & Science, Carlsbad, California, USA
| | | | | | | | | | | | | | | | - Ke K. Zhang
- Bioinformatics core, Department of Pathology, University of North Dakota, Grand Forks, North Dakota, USA
| | - Jianying Li
- Microarray and Genome Informatics Group, National Institute of Environmental Health Sciences, RTP, North Carolina, USA
- Kelly Government Solutions, Inc., Durham, North Carolina, USA
| | - Jui-Hua Hsieh
- Biomolecular Screening Branch, Division of the National Toxicology Program, National Institute of Environmental Health Sciences, RTP, North Carolina, USA
| | | | - James C. Fuscoe
- Division of Systems Biology, National Center for Toxicological Research, US Food and Drug Administration, Jefferson, Arkansas, USA
| | - Youping Deng
- Department of Internal Medicine and Biochemistry, Rush University Medical Center, Chicago, Illinois, USA
| | - Leming Shi
- Division of Bioinformatics and Biostatistics, National Center for Toxicological Research, US Food and Drug Administration, Jefferson, Arkansas, USA
- Center for Pharmacogenomics, Schools of Life Sciences and Pharmacy, Fudan University, Shanghai, China
| | - Richard S. Paules
- Laboratory of Toxicology and Pharmacology, National Institute of Environmental Health Sciences, RTP, North Carolina, USA
| | - Scott S. Auerbach
- Biomolecular Screening Branch, Division of the National Toxicology Program, National Institute of Environmental Health Sciences, RTP, North Carolina, USA
| | - Weida Tong
- Division of Bioinformatics and Biostatistics, National Center for Toxicological Research, US Food and Drug Administration, Jefferson, Arkansas, USA
| |
Collapse
|
172
|
Wang K, Zhao L, Liu X, Hao Z, Zhou Y, Yang C, Li H. Differential co-expression analysis of rheumatoid arthritis with microarray data. Mol Med Rep 2014; 10:2421-6. [PMID: 25118911 DOI: 10.3892/mmr.2014.2491] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2013] [Accepted: 04/25/2014] [Indexed: 11/06/2022] Open
Abstract
The aim of the present study was to investigate the underlying molecular mechanisms of rheumatoid arthritis (RA) using microarray expression profiles from osteoarthritis and RA patients, to improve diagnosis and treatment strategies for the condition. The gene expression profile of GSE27390 was downloaded from Gene Expression Omnibus, including 19 samples from patients with RA (n=9) or osteoarthritis (n=10). Firstly, the differentially expressed genes (DEGs) were obtained with the thresholds of |logFC|>1.0 and P<0.05, using the t‑test method in LIMMA package. Then, differentially co-expressed genes (DCGs) and differentially co-expressed links (DCLs) were screened with q<0.25 by the differential coexpression analysis and differential regulation analysis of gene expression microarray data package. Secondly, pathway enrichment analysis for DCGs was performed by the Database for Annotation, Visualization and Integrated Discovery and the DCLs associated with RA were selected by comparing the obtained DCLs with known transcription factor (TF)-targets in the TRANSFAC database. Finally, the obtained TFs were mapped to the known TF-targets to construct the network using cytoscape software. A total of 1755 DEGs, 457 DCGs and 101988 DCLs were achieved and there were 20 TFs in the obtained six TF-target relations (STAT3-TNF, PBX1‑PLAU, SOCS3-STAT3, GATA1-ETS2, ETS1-ICAM4 and CEBPE‑GATA1) and 457 DCGs. A number of TF-target relations in the constructed network were not within DCLs when the TF and target gene were DCGs. The identified TFs may have an important role in the pathogenesis of RA and have the potential to be used as biomarkers for the development of novel diagnostic and therapeutic strategies for RA.
Collapse
Affiliation(s)
- Kunpeng Wang
- Department of Orthopedics, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Liqiang Zhao
- Department of Orthopedics, The Harbin Fifth Hospital, Harbin, Heilongjiang 150001, P.R. China
| | - Xuefeng Liu
- Department of Orthopedics, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Zhenyong Hao
- Department of Orthopedics, The Harbin Fifth Hospital, Harbin, Heilongjiang 150001, P.R. China
| | - Yong Zhou
- Department of Orthopedics, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Chuandong Yang
- Department of Orthopedics, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Hongqiang Li
- Department of Orthopedics, The Harbin Fifth Hospital, Harbin, Heilongjiang 150001, P.R. China
| |
Collapse
|
173
|
Faisal M, Waseem D, Ismatullah H, Taqi MM. A molecular prospective provides new insights into implication of PDYN and OPRK1 genes in alcohol dependence. Comput Biol Med 2014; 53:250-7. [PMID: 25177835 DOI: 10.1016/j.compbiomed.2014.07.021] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2013] [Revised: 07/24/2014] [Accepted: 07/26/2014] [Indexed: 12/15/2022]
Abstract
Single nucleotide polymorphisms (SNPs) both in coding and non-coding regions govern gene functions prompting differential vulnerability to diseases, heterogeneous response to pharmaceutical regimes and environmental anomalies. These genetic variations, SNPs, may alter an individual׳s susceptibility for alcohol dependence by remodeling DNA-protein interaction patterns in prodynorphin (PDYN) and the κ-opioid receptor (OPRK1) genes. In order to elaborate the underlying molecular mechanism behind these susceptibility differences we used bioinformatics tools to retrieve differential DNA-protein interactions at PDYN and OPRK1 SNPs significantly associated with alcohol dependence. Our results show allele-specific DNA-protein interactions depicting allele-specific mechanisms implicated in differential regulation of gene expression. Several transcription factors, for instance, VDR, RXR-alpha, NFYA, CTF family, USF-1, USF2, ER, AR and predominantly SP family show an allele-specific binding affinity with PDYN gene; likewise, GATA, TBP, AP-1, USF-2, C/EBPbeta, Cart-1 and ER interact with OPRK1 SNPs on intron 2 in an allele-specific manner. In a nutshell, transition of a single nucleotide may modify differential DNA-protein interactions at OPRK1 and PDYN׳s SNPs, significantly associated with pathology that may lead to altered individual vulnerability for alcohol dependence.
Collapse
Affiliation(s)
- Muhammad Faisal
- National Center for Bioinformatics, Quaid-i-Azam University, Islamabad, Pakistan
| | - Durdana Waseem
- Department of Pharmacy, Quaid-i-Azam University, Islamabad 45320, Pakistan
| | - Humaira Ismatullah
- Department of Bioinformatics and Biotechnology, International Islamic University, Islamabad, Pakistan
| | - Malik Mumtaz Taqi
- Department of Pharmacy, Quaid-i-Azam University, Islamabad 45320, Pakistan.
| |
Collapse
|
174
|
Backes C, Rühle F, Stoll M, Haas J, Frese K, Franke A, Lieb W, Wichmann HE, Weis T, Kloos W, Lenhof HP, Meese E, Katus H, Meder B, Keller A. Systematic permutation testing in GWAS pathway analyses: identification of genetic networks in dilated cardiomyopathy and ulcerative colitis. BMC Genomics 2014; 15:622. [PMID: 25052024 PMCID: PMC4223581 DOI: 10.1186/1471-2164-15-622] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2014] [Accepted: 07/17/2014] [Indexed: 12/20/2022] Open
Abstract
Background Genome wide association studies (GWAS) are applied to identify genetic loci, which are associated with complex traits and human diseases. Analogous to the evolution of gene expression analyses, pathway analyses have emerged as important tools to uncover functional networks of genome-wide association data. Usually, pathway analyses combine statistical methods with a priori available biological knowledge. To determine significance thresholds for associated pathways, correction for multiple testing and over-representation permutation testing is applied. Results We systematically investigated the impact of three different permutation test approaches for over-representation analysis to detect false positive pathway candidates and evaluate them on genome-wide association data of Dilated Cardiomyopathy (DCM) and Ulcerative Colitis (UC). Our results provide evidence that the gold standard - permuting the case–control status – effectively improves specificity of GWAS pathway analysis. Although permutation of SNPs does not maintain linkage disequilibrium (LD), these permutations represent an alternative for GWAS data when case–control permutations are not possible. Gene permutations, however, did not add significantly to the specificity. Finally, we provide estimates on the required number of permutations for the investigated approaches. Conclusions To discover potential false positive functional pathway candidates and to support the results from standard statistical tests such as the Hypergeometric test, permutation tests of case control data should be carried out. The most reasonable alternative was case–control permutation, if this is not possible, SNP permutations may be carried out. Our study also demonstrates that significance values converge rapidly with an increasing number of permutations. By applying the described statistical framework we were able to discover axon guidance, focal adhesion and calcium signaling as important DCM-related pathways and Intestinal immune network for IgA production as most significant UC pathway.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | - Andreas Keller
- Chair for Clinical Bioinformatics, Saarland University, Saarbrücken, Germany.
| |
Collapse
|
175
|
Leung A, Parks BW, Du J, Trac C, Setten R, Chen Y, Brown K, Lusis AJ, Natarajan R, Schones DE. Open chromatin profiling in mice livers reveals unique chromatin variations induced by high fat diet. J Biol Chem 2014; 289:23557-67. [PMID: 25006255 DOI: 10.1074/jbc.m114.581439] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Metabolic diseases result from multiple genetic and environmental factors. We report here that one manner in which environmental factors can contribute to metabolic disease progression is through modification to chromatin. We demonstrate that high fat diet leads to chromatin remodeling in the livers of C57BL/6J mice, as compared with mice fed a control diet, and that these chromatin changes are associated with changes in gene expression. We further show that the regions of greatest variation in chromatin accessibility are targeted by liver transcription factors, including HNF4α, CCAAT/enhancer-binding protein α (CEBP/α), and FOXA1. Repeating the chromatin and gene expression profiling in another mouse strain, DBA/2J, revealed that the regions of greatest chromatin change are largely strain-specific and that integration of chromatin, gene expression, and genetic data can be used to characterize regulatory regions. Our data indicate dramatic changes in the epigenome due to diet and demonstrate strain-specific dynamics in chromatin remodeling.
Collapse
Affiliation(s)
- Amy Leung
- From the Departments of Diabetes and
| | - Brian W Parks
- the Department of Medicine, UCLA, Los Angeles, California 90095
| | - Juan Du
- Cancer Biology, Beckman Research Institute and the Irell & Manella Graduate School of Biological Sciences, City of Hope, Duarte, California 91010 and
| | - Candi Trac
- Cancer Biology, Beckman Research Institute and
| | - Ryan Setten
- the Irell & Manella Graduate School of Biological Sciences, City of Hope, Duarte, California 91010 and
| | - Yin Chen
- the Irell & Manella Graduate School of Biological Sciences, City of Hope, Duarte, California 91010 and
| | - Kevin Brown
- Cancer Biology, Beckman Research Institute and
| | - Aldons J Lusis
- the Department of Medicine, UCLA, Los Angeles, California 90095
| | - Rama Natarajan
- From the Departments of Diabetes and the Irell & Manella Graduate School of Biological Sciences, City of Hope, Duarte, California 91010 and
| | - Dustin E Schones
- Cancer Biology, Beckman Research Institute and the Irell & Manella Graduate School of Biological Sciences, City of Hope, Duarte, California 91010 and
| |
Collapse
|
176
|
Papanayotou C, Benhaddou A, Camus A, Perea-Gomez A, Jouneau A, Mezger V, Langa F, Ott S, Sabéran-Djoneidi D, Collignon J. A novel nodal enhancer dependent on pluripotency factors and smad2/3 signaling conditions a regulatory switch during epiblast maturation. PLoS Biol 2014; 12:e1001890. [PMID: 24960041 PMCID: PMC4068991 DOI: 10.1371/journal.pbio.1001890] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2013] [Accepted: 05/15/2014] [Indexed: 02/07/2023] Open
Abstract
HBE, a newly discovered enhancer element, mediates the influence of pluripotency factors and Activin/Nodal signaling on early Nodal expression in the mouse embryo, and controls the activation of later-acting Nodal enhancers. During early development, modulations in the expression of Nodal, a TGFβ family member, determine the specification of embryonic and extra-embryonic cell identities. Nodal has been extensively studied in the mouse, but aspects of its early expression remain unaccounted for. We identified a conserved hotspot for the binding of pluripotency factors at the Nodal locus and called this sequence “highly bound element” (HBE). Luciferase-based assays, the analysis of fluorescent HBE reporter transgenes, and a conditional mutation of HBE allowed us to establish that HBE behaves as an enhancer, is activated ahead of other Nodal enhancers in the epiblast, and is essential to Nodal expression in embryonic stem cells (ESCs) and in the mouse embryo. We also showed that HBE enhancer activity is critically dependent on its interaction with the pluripotency factor Oct4 and on Activin/Nodal signaling. Use of an in vitro model of epiblast maturation, relying on the differentiation of ESCs into epiblast stem cells (EpiSCs), revealed that this process entails a shift in the regulation of Nodal expression from an HBE-driven phase to an ASE-driven phase, ASE being another autoregulatory Nodal enhancer. Deletion of HBE in ESCs or in EpiSCs allowed us to show that HBE, although not necessary for Nodal expression in EpiSCs, is required in differentiating ESCs to activate the differentiation-promoting ASE and therefore controls this regulatory shift. Our findings clarify how early Nodal expression is regulated and suggest how this regulation can promote the specification of extra-embryonic precusors without inducing premature differentiation of epiblast cells. More generally, they open new perspectives on how pluripotency factors achieve their function. In the early mouse embryo, Nodal, a member of the TGFbeta superfamily of signalling proteins, promotes the differentiation of extra-embryonic tissues, as well as tissues within the developing embryo itself. Characterising the regulation of Nodal gene expression is essential to understand how Nodal signals in diverse tissue types and at different stages of embryonic development. Four distinct enhancer sequences have been shown to regulate Nodal expression, although none could account for it in the preimplantation epiblast or in embryonic stem cells. We identified a novel enhancer, HBE, responsible for the earliest aspects of Nodal expression. We show that activation of HBE depends on its interaction with a well-known pluripotency factor called Oct4. HBE itself also controls the activation of at least one other Nodal enhancer. Our findings clarify how early Nodal expression is regulated and reveal how pluripotency factors may control the onset of differentiation in embryonic tissues.
Collapse
Affiliation(s)
- Costis Papanayotou
- Institut Jacques Monod, UMR 7592, CNRS, Université Paris-Diderot, Sorbonne Paris Cité, Paris, France
- * E-mail: (JC); (CP)
| | - Ataaillah Benhaddou
- Institut Jacques Monod, UMR 7592, CNRS, Université Paris-Diderot, Sorbonne Paris Cité, Paris, France
| | - Anne Camus
- Institut Jacques Monod, UMR 7592, CNRS, Université Paris-Diderot, Sorbonne Paris Cité, Paris, France
| | - Aitana Perea-Gomez
- Institut Jacques Monod, UMR 7592, CNRS, Université Paris-Diderot, Sorbonne Paris Cité, Paris, France
| | - Alice Jouneau
- Unité de Biologie du Développement et de la reproduction, UMR INRA-ENVA, INRA, Jouy-en-Josas, France
| | - Valérie Mezger
- Epigenetics and Cell Fate, UMR7216, CNRS, Université Paris-Diderot, Sorbonne Paris Cité, Paris, France
| | - Francina Langa
- Centre d'Ingénierie Génétique Murine, Institut Pasteur, Paris, France
| | - Sascha Ott
- Warwick Systems Biology Centre, University of Warwick, Coventry, United Kingdom
| | - Délara Sabéran-Djoneidi
- Institut Jacques Monod, UMR 7592, CNRS, Université Paris-Diderot, Sorbonne Paris Cité, Paris, France
- Epigenetics and Cell Fate, UMR7216, CNRS, Université Paris-Diderot, Sorbonne Paris Cité, Paris, France
| | - Jérôme Collignon
- Institut Jacques Monod, UMR 7592, CNRS, Université Paris-Diderot, Sorbonne Paris Cité, Paris, France
- * E-mail: (JC); (CP)
| |
Collapse
|
177
|
Fu S, Pan X, Fang W. Differential co-expression analysis of a microarray gene expression profiles of pulmonary adenocarcinoma. Mol Med Rep 2014; 10:713-8. [PMID: 24913908 DOI: 10.3892/mmr.2014.2300] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2013] [Accepted: 03/17/2014] [Indexed: 11/06/2022] Open
Abstract
Lung cancer severely reduces the quality of life worldwide and causes high socioeconomic burdens. However, key genes leading to the generation of pulmonary adenocarcinoma remain elusive despite intensive research efforts. The present study aimed to identify the potential associations between transcription factors (TFs) and differentially co‑expressed genes (DCGs) in the regulation of transcription in pulmonary adenocarcinoma. Gene expression profiles of pulmonary adenocarcinoma were downloaded from the Gene Expression Omnibus, and gene expression was analyzed using a computational method. A total of 37,094 differentially co‑expressed links (DCLs) and 251 DCGs were identified, which were significantly enriched in 10 pathways. The construction of the regulatory network and the analysis of the regulatory impact factors revealed eight crucial TFs in the regulatory network. These TFs regulated the expression of DCGs by promoting or inhibiting their expression. In addition, certain TFs and target genes associated with DCGs did not appear in the DCLs, which indicated that those TFs could be synergistic with other factors. This is likely to provide novel insights for research into pulmonary adenocarcinoma. In conclusion, the present study may enhance the understanding of disease mechanisms and lead to an improved diagnosis of lung cancer. However, further studies are required to confirm these observations.
Collapse
Affiliation(s)
- Shijie Fu
- Department of Thoracic Surgery, Shanghai Chest Hospital, Shanghai Jiaotong University, Shanghai 200030, P.R. China
| | - Xufeng Pan
- Department of Thoracic Surgery, Shanghai Chest Hospital, Shanghai Jiaotong University, Shanghai 200030, P.R. China
| | - Wentao Fang
- Department of Thoracic Surgery, Shanghai Chest Hospital, Shanghai Jiaotong University, Shanghai 200030, P.R. China
| |
Collapse
|
178
|
Pathway and network analysis in proteomics. J Theor Biol 2014; 362:44-52. [PMID: 24911777 DOI: 10.1016/j.jtbi.2014.05.031] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2014] [Revised: 05/15/2014] [Accepted: 05/21/2014] [Indexed: 12/14/2022]
Abstract
Proteomics is inherently a systems science that studies not only measured protein and their expressions in a cell, but also the interplay of proteins, protein complexes, signaling pathways, and network modules. There is a rapid accumulation of Proteomics data in recent years. However, Proteomics data are highly variable, with results sensitive to data preparation methods, sample condition, instrument types, and analytical methods. To address the challenge in Proteomics data analysis, we review current tools being developed to incorporate biological function and network topological information. We categorize these tools into four types: tools with basic functional information and little topological features (e.g., GO category analysis), tools with rich functional information and little topological features (e.g., GSEA), tools with basic functional information and rich topological features (e.g., Cytoscape), and tools with rich functional information and rich topological features (e.g., PathwayExpress). We first review the potential application of these tools to Proteomics; then we review tools that can achieve automated learning of pathway modules and features, and tools that help perform integrated network visual analytics.
Collapse
|
179
|
Li Y, Liang C, Wong KC, Luo J, Zhang Z. Mirsynergy: detecting synergistic miRNA regulatory modules by overlapping neighbourhood expansion. ACTA ACUST UNITED AC 2014; 30:2627-35. [PMID: 24894504 DOI: 10.1093/bioinformatics/btu373] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
MOTIVATION Identification of microRNA regulatory modules (MiRMs) will aid deciphering aberrant transcriptional regulatory network in cancer but is computationally challenging. Existing methods are stochastic or require a fixed number of regulatory modules. RESULTS We propose Mirsynergy, an efficient deterministic overlapping clustering algorithm adapted from a recently developed framework. Mirsynergy operates in two stages: it first forms MiRMs based on co-occurring microRNA (miRNA) targets and then expands each MiRM by greedily including (excluding) mRNAs into (from) the MiRM to maximize the synergy score, which is a function of miRNA-mRNA and gene-gene interactions. Using expression data for ovarian, breast and thyroid cancer from The Cancer Genome Atlas, we compared Mirsynergy with internal controls and existing methods. Mirsynergy-MiRMs exhibit significantly higher functional enrichment and more coherent miRNA-mRNA expression anti-correlation. Based on Kaplan-Meier survival analysis, we proposed several prognostically promising MiRMs and envisioned their utility in cancer research. AVAILABILITY AND IMPLEMENTATION Mirsynergy is implemented/available as an R/Bioconductor package at www.cs.utoronto.ca/∼yueli/Mirsynergy.html.
Collapse
Affiliation(s)
- Yue Li
- Department of Computer Science, The Donnelly Centre, University of Toronto, Toronto, Ontario M5S 3E1, Canada, College of Information Science and Engineering, Hunan University, Changsha, Hunan, 410082, China and Banting and Best Department of Medical Research and Department of Molecular Genetics, University of Toronto, Toronto, Ontario M5S 3E1, Canada Department of Computer Science, The Donnelly Centre, University of Toronto, Toronto, Ontario M5S 3E1, Canada, College of Information Science and Engineering, Hunan University, Changsha, Hunan, 410082, China and Banting and Best Department of Medical Research and Department of Molecular Genetics, University of Toronto, Toronto, Ontario M5S 3E1, Canada
| | - Cheng Liang
- Department of Computer Science, The Donnelly Centre, University of Toronto, Toronto, Ontario M5S 3E1, Canada, College of Information Science and Engineering, Hunan University, Changsha, Hunan, 410082, China and Banting and Best Department of Medical Research and Department of Molecular Genetics, University of Toronto, Toronto, Ontario M5S 3E1, Canada
| | - Ka-Chun Wong
- Department of Computer Science, The Donnelly Centre, University of Toronto, Toronto, Ontario M5S 3E1, Canada, College of Information Science and Engineering, Hunan University, Changsha, Hunan, 410082, China and Banting and Best Department of Medical Research and Department of Molecular Genetics, University of Toronto, Toronto, Ontario M5S 3E1, Canada Department of Computer Science, The Donnelly Centre, University of Toronto, Toronto, Ontario M5S 3E1, Canada, College of Information Science and Engineering, Hunan University, Changsha, Hunan, 410082, China and Banting and Best Department of Medical Research and Department of Molecular Genetics, University of Toronto, Toronto, Ontario M5S 3E1, Canada
| | - Jiawei Luo
- Department of Computer Science, The Donnelly Centre, University of Toronto, Toronto, Ontario M5S 3E1, Canada, College of Information Science and Engineering, Hunan University, Changsha, Hunan, 410082, China and Banting and Best Department of Medical Research and Department of Molecular Genetics, University of Toronto, Toronto, Ontario M5S 3E1, Canada
| | - Zhaolei Zhang
- Department of Computer Science, The Donnelly Centre, University of Toronto, Toronto, Ontario M5S 3E1, Canada, College of Information Science and Engineering, Hunan University, Changsha, Hunan, 410082, China and Banting and Best Department of Medical Research and Department of Molecular Genetics, University of Toronto, Toronto, Ontario M5S 3E1, Canada Department of Computer Science, The Donnelly Centre, University of Toronto, Toronto, Ontario M5S 3E1, Canada, College of Information Science and Engineering, Hunan University, Changsha, Hunan, 410082, China and Banting and Best Department of Medical Research and Department of Molecular Genetics, University of Toronto, Toronto, Ontario M5S 3E1, Canada Department of Computer Science, The Donnelly Centre, University of Toronto, Toronto, Ontario M5S 3E1, Canada, College of Information Science and Engineering, Hunan University, Changsha, Hunan, 410082, China and Banting and Best Department of Medical Research and Department of Molecular Genetics, University of Toronto, Toronto, Ontario M5S 3E1, Canada
| |
Collapse
|
180
|
Downarowicz T, Travisany D, Montecino M, Maass A. Symbolic extensions applied to multiscale structure of genomes. Acta Biotheor 2014; 62:145-69. [PMID: 24728912 PMCID: PMC4012164 DOI: 10.1007/s10441-014-9215-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2013] [Accepted: 03/25/2014] [Indexed: 11/27/2022]
Abstract
A genome of a living organism consists of a long string of symbols over a finite alphabet carrying critical information for the organism. This includes its ability to control post natal growth, homeostasis, adaptation to changes in the surrounding environment, or to biochemically respond at the cellular level to various specific regulatory signals. In this sense, a genome represents a symbolic encoding of a highly organized system of information whose functioning may be revealed as a natural multilayer structure in terms of complexity and prominence. In this paper we use the mathematical theory of symbolic extensions as a framework to shed light onto how this multilayer organization is reflected in the symbolic coding of the genome. The distribution of data in an element of a standard symbolic extension of a dynamical system has a specific form: the symbolic sequence is divided into several subsequences (which we call layers) encoding the dynamics on various "scales". We propose that a similar structure resides within the genomes, building our analogy on some of the most recent findings in the field of regulation of genomic DNA functioning.
Collapse
Affiliation(s)
- Tomasz Downarowicz
- Institute of Mathematics and Computer Science, Wroclaw University of Technology, Wybrzeże Wyspiańskiego 27, 50-370 Wrocław, Poland
| | - Dante Travisany
- Center for Mathematical Modeling, FONDAP Center for Genome Regulation, University of Chile, Blanco Encalada 2120, Santiago, Chile
| | - Martin Montecino
- Faculty of Biological Sciences and Faculty of Medicine, Center for Biomedical Research, FONDAP Center for Genome Regulation, Universidad Andrés Bello, Avenida República 239, Santiago, Chile
| | - Alejandro Maass
- Department of Mathematical Engineering, Center for Mathematical Modeling, FONDAP Center for Genome Regulation, University of Chile, Blanco Encalada 2120, Santiago, Chile
| |
Collapse
|
181
|
Kim K, Ban HJ, Seo J, Lee K, Yavartanoo M, Kim SC, Park K, Cho SB, Choi JK. Genetic factors underlying discordance in chromatin accessibility between monozygotic twins. Genome Biol 2014; 15:R72. [PMID: 24887574 PMCID: PMC4072931 DOI: 10.1186/gb-2014-15-5-r72] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2013] [Accepted: 05/29/2014] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Open chromatin is implicated in regulatory processes; thus, variations in chromatin structure may contribute to variations in gene expression and other phenotypes. In this work, we perform targeted deep sequencing for open chromatin, and array-based genotyping across the genomes of 72 monozygotic twins to identify genetic factors regulating co-twin discordance in chromatin accessibility. RESULTS We show that somatic mutations cause chromatin discordance mainly via the disruption of transcription factor binding sites. Structural changes in DNA due to C:G to A:T transversions are under purifying selection due to a strong impact on chromatin accessibility. We show that CpGs whose methylation is specifically regulated during cellular differentiation appear to be protected from high mutation rates of 5'-methylcytosines, suggesting that the spectrum of CpG variations may be shaped fully at the developmental level but not through natural selection. Based on the association mapping of within-pair chromatin differences, we search for cases in which twin siblings with a particular genotype had chromatin discordance at the relevant locus. We identify 1,325 chromatin sites that are differentially accessible, depending on the genotype of a nearby locus, suggesting that epigenetic differences can control regulatory variations via interactions with genetic factors. Poised promoters present high levels of chromatin discordance in association with either somatic mutations or genetic-epigenetic interactions. CONCLUSION Our observations illustrate how somatic mutations and genetic polymorphisms may contribute to regulatory, and ultimately phenotypic, discordance.
Collapse
|
182
|
Bosquet JG, Marchion DC, Chon H, Lancaster JM, Chanock S. Analysis of chemotherapeutic response in ovarian cancers using publicly available high-throughput data. Cancer Res 2014; 74:3902-12. [PMID: 24848511 DOI: 10.1158/0008-5472.can-14-0186] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
A third of patients with epithelial ovarian cancer (OVCA) will not respond to standard treatment. The determination of a robust signature that predicts chemoresponse could lead to the identification of molecular markers for response as well as possible clinical implementation in the future to identify patients at risk of failing therapy. This pilot study was designed to identify biologic processes affecting candidate pathways associated with chemoresponse and to create a robust gene signature for follow-up studies. After identifying common pathways associated with chemoresponse in serous OVCA in three independent gene-expression experiments, we assessed the biologic processes associated with them using The Cancer Genome Atlas (TCGA) dataset for serous OVCA. We identified differential copy-number alterations (CNA), mutations, DNA methylation, and miRNA expression between patients that responded to standard treatment and those who did not or recurred prematurely. We correlated these significant parameters with gene expression to create a signature of 422 genes associated with chemoresponse. A consensus clustering of this signature identified two differentiated clusters with unique molecular patterns: cluster 1 was significant for cellular signaling and immune response (mainly cell-mediated); and cluster 2 was significant for pathways involving DNA-damage repair and replication, cell cycle, and apoptosis. Validation through consensus clustering was performed in five independent OVCA gene-expression experiments. Genes were located in the same cluster with consistent agreement in all five studies (κ coefficient ≥ 0.6 in 4). Integrating high-throughput biologic data have created a robust molecular signature that predicts chemoresponse in OVCA.
Collapse
Affiliation(s)
- Jesus Gonzalez Bosquet
- Authors' Affiliations: Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of Iowa Hospitals and Clinics, Iowa City, Iowa;
| | - Douglas C Marchion
- Department of Women's Oncology, Experimental Therapeutics Program, Department of Oncologic Sciences
| | - HyeSook Chon
- Department of Women's Oncology, Gynecologic Oncology, Department of Oncologic Sciences, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida; and
| | - Johnathan M Lancaster
- Department of Women's Oncology, Gynecologic Oncology, Department of Oncologic Sciences, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida; and
| | - Stephen Chanock
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Bethesda, Maryland
| |
Collapse
|
183
|
Mohanta R, Jayasankar P, Das Mahapatra K, Saha JN, Barman HK. Molecular cloning, characterization and functional assessment of the myosin light polypeptide chain 2 (mylz2) promoter of farmed carp, Labeo rohita. Transgenic Res 2014; 23:601-7. [PMID: 24740361 DOI: 10.1007/s11248-014-9798-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2014] [Accepted: 04/03/2014] [Indexed: 01/07/2023]
Abstract
We cloned the 5'-flanking region (1.2 kb) of a muscle-specific gene, encoding myosin light chain 2 polypeptide (mylz2) of a farmed carp, Labeo rohita (rohu). Sequence analysis using TRANSFAC-database search identified the consensus cis acting regulatory elements of TATA-box and E (CANNTG)-box, including the monocyte enhancer factor 2 motif, implying that it is likely to be a functional promoter. The proximal promoter (~620 bp) was highly homologous with that of Danio rerio (zebrafish) as compared to Channa striatus (snakehead murrel) counterparts and showed less identity with Sparus auratus (gilthead sea bream), Xenopus laevis (African clawed frog) and Rattus norvegicus (Norway rat). Direct muscular (skeletal) injection of the construct containing the mylz2 promoter (0.6 kb) fused to a green fluorescent protein (GFP) reporter gene showed efficient expression in L. rohita, validating its functional activity. Further, the functional activity was confirmed by the observation that this promoter drove GFP expression in the skeletal muscle of transgenic rohu. The promoter may have potential applications for value-addition in ornamental fishes and studying gene regulatory functions.
Collapse
Affiliation(s)
- Ramya Mohanta
- Fish Genetics and Biotechnology Division, Central Institute of Freshwater Aquaculture (Indian Council of Agricultural Research), Kausalyaganga, Bhubaneswar, 751002, Odisha, India
| | | | | | | | | |
Collapse
|
184
|
Park SJ, Umemoto T, Saito-Adachi M, Shiratsuchi Y, Yamato M, Nakai K. Computational promoter modeling identifies the modes of transcriptional regulation in hematopoietic stem cells. PLoS One 2014; 9:e93853. [PMID: 24710559 PMCID: PMC3977923 DOI: 10.1371/journal.pone.0093853] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2013] [Accepted: 03/07/2014] [Indexed: 01/19/2023] Open
Abstract
Extrinsic and intrinsic regulators are responsible for the tight control of hematopoietic stem cells (HSCs), which differentiate into all blood cell lineages. To understand the fundamental basis of HSC biology, we focused on differentially expressed genes (DEGs) in long-term and short-term HSCs, which are closely related in terms of cell development but substantially differ in their stem cell capacity. To analyze the transcriptional regulation of the DEGs identified in the novel transcriptome profiles obtained by our RNA-seq analysis, we developed a computational method to model the linear relationship between gene expression and the features of putative regulatory elements. The transcriptional regulation modes characterized here suggest the importance of transcription factors (TFs) that are expressed at steady state or at low levels. Remarkably, we found that 24 differentially expressed TFs targeting 21 putative TF-binding sites contributed significantly to transcriptional regulation. These TFs tended to be modulated by other nondifferentially expressed TFs, suggesting that HSCs can achieve flexible and rapid responses via the control of nondifferentially expressed TFs through a highly complex regulatory network. Our novel transcriptome profiles and new method are powerful tools for studying the mechanistic basis of cell fate decisions.
Collapse
Affiliation(s)
- Sung-Joon Park
- Human Genome Center, the Institute of Medical Science, the University of Tokyo, Tokyo, Japan
| | - Terumasa Umemoto
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University, Tokyo, Japan
| | - Mihoko Saito-Adachi
- Human Genome Center, the Institute of Medical Science, the University of Tokyo, Tokyo, Japan
| | - Yoshiko Shiratsuchi
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University, Tokyo, Japan
| | - Masayuki Yamato
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University, Tokyo, Japan
| | - Kenta Nakai
- Human Genome Center, the Institute of Medical Science, the University of Tokyo, Tokyo, Japan
| |
Collapse
|
185
|
Abstract
Efforts to understand the molecular basis of mycobacterial gene regulation are dominated by a protein-centric view. However, there is a growing appreciation that noncoding RNA, i.e., RNA that is not translated, plays a role in a wide variety of molecular mechanisms. Noncoding RNA comprises rRNA, tRNA, 4.5S RNA, RnpB, and transfer-messenger RNA, as well as a vast population of regulatory RNA, often dubbed "the dark matter of gene regulation." The regulatory RNA species comprise 5' and 3' untranslated regions and a rapidly expanding category of transcripts with the ability to base-pair with mRNAs or to interact with proteins. Regulatory RNA plays a central role in the bacterium's response to changes in the environment, and in this article we review emerging information on the presence and abundance of different types of noncoding RNA in mycobacteria.
Collapse
|
186
|
Sastre-Perona A, Santisteban P. Wnt-independent role of β-catenin in thyroid cell proliferation and differentiation. Mol Endocrinol 2014; 28:681-95. [PMID: 24645679 DOI: 10.1210/me.2013-1377] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The Wnt/β-catenin pathway has been associated with thyroid cell growth and tumorigenesis. However, little is known regarding its involvement in the response to the key regulators of thyroid cell proliferation and differentiation. Here we show that TSH and IGF-1 increase β-catenin nuclear accumulation and its transcriptional activity in differentiated thyroid cells. This effect takes place in a Wnt-independent manner because TSH and IGF-1, through the activation of protein kinase A and protein kinase B/Akt, phosphorylate β-catenin at S552 and S675, which results in β-catenin release from E-cadherin at the adherens junctions. Nuclear β-catenin regulates thyroid cell proliferation, because its silencing or the overexpression of a dominant-negative form of T-cell factor 4 resulted in reduced levels of cyclin D1 and DNA synthesis. Furthermore, the β-catenin silencing markedly reduced the expression of Pax8, the main transcription factor involved in epithelial thyroid cell differentiation. Finally, we observed that β-catenin physically interacts with the transcription factor Pax8, increasing its transcriptional activity on the sodium iodide symporter (NIS) gene, a critical gene required for thyroid cell physiology. Taken together, our findings show that β-catenin plays a not yet described role in thyroid function including a functional interaction with Pax8.
Collapse
Affiliation(s)
- Ana Sastre-Perona
- Instituto de Investigaciones Biomédicas "Alberto Sols" Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid, 28029, Madrid, Spain
| | | |
Collapse
|
187
|
Neurodevelopmental and neuropsychiatric disorders represent an interconnected molecular system. Mol Psychiatry 2014; 19:294-301. [PMID: 23439483 DOI: 10.1038/mp.2013.16] [Citation(s) in RCA: 147] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2012] [Revised: 12/14/2012] [Accepted: 01/02/2013] [Indexed: 12/18/2022]
Abstract
Many putative genetic factors that confer risk to neurodevelopmental disorders such as autism spectrum disorders (ASDs) and X-linked intellectual disability (XLID), and to neuropsychiatric disorders including attention deficit hyperactivity disorder (ADHD) and schizophrenia (SZ) have been identified in individuals from diverse human populations. Although there is significant aetiological heterogeneity within and between these conditions, recent data show that genetic factors contribute to their comorbidity. Many studies have identified candidate gene associations for these mental health disorders, albeit this is often done in a piecemeal fashion with little regard to the inherent molecular complexity. Here, we sought to abstract relationships from our knowledge of systems level biology to help understand the unique and common genetic drivers of these conditions. We undertook a global and systematic approach to build and integrate available data in gene networks associated with ASDs, XLID, ADHD and SZ. Complex network concepts and computational methods were used to investigate whether candidate genes associated with these conditions were related through mechanisms of gene regulation, functional protein-protein interactions, transcription factor (TF) and microRNA (miRNA) binding sites. Although our analyses show that genetic variations associated with the four disorders can occur in the same molecular pathways and functional domains, including synaptic transmission, there are patterns of variation that define significant differences between disorders. Of particular interest is DNA variations located in intergenic regions that comprise regulatory sites for TFs or miRNA. Our approach provides a hypothetical framework, which will help discovery and analysis of candidate genes associated with neurodevelopmental and neuropsychiatric disorders.
Collapse
|
188
|
Bateman AR, El-Hachem N, Beck AH, Aerts HJWL, Haibe-Kains B. Importance of collection in gene set enrichment analysis of drug response in cancer cell lines. Sci Rep 2014; 4:4092. [PMID: 24522610 PMCID: PMC3923229 DOI: 10.1038/srep04092] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2013] [Accepted: 01/29/2014] [Indexed: 12/27/2022] Open
Abstract
Gene set enrichment analysis (GSEA) associates gene sets and phenotypes, its use is predicated on the choice of a pre-defined collection of sets. The defacto standard implementation of GSEA provides seven collections yet there are no guidelines for the choice of collections and the impact of such choice, if any, is unknown. Here we compare each of the standard gene set collections in the context of a large dataset of drug response in human cancer cell lines. We define and test a new collection based on gene co-expression in cancer cell lines to compare the performance of the standard collections to an externally derived cell line based collection. The results show that GSEA findings vary significantly depending on the collection chosen for analysis. Henceforth, collections should be carefully selected and reported in studies that leverage GSEA.
Collapse
Affiliation(s)
- Alain R Bateman
- Bioinformatics and Computational Genomics Laboratory, Institut de Recherches Cliniques de Montréal, University of Montreal, Montreal, Quebec, Canada
| | - Nehme El-Hachem
- Bioinformatics and Computational Genomics Laboratory, Institut de Recherches Cliniques de Montréal, University of Montreal, Montreal, Quebec, Canada
| | - Andrew H Beck
- Department of Pathology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Hugo J W L Aerts
- 1] Department of Biostatistics and Computational Biology and Center for Cancer Computational Biology, Dana-Farber Cancer Institute, Boston, MA, USA [2] Department of Radiation Oncology & Radiology, Dana-Farber Cancer Institute, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA [3] Department of Radiation Oncology, Maastricht University, Maastricht, The Netherlands
| | - Benjamin Haibe-Kains
- 1] Bioinformatics and Computational Genomics Laboratory, Institut de Recherches Cliniques de Montréal, University of Montreal, Montreal, Quebec, Canada [2] Ontario Cancer Institute, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| |
Collapse
|
189
|
Park SJ, Komata M, Inoue F, Yamada K, Nakai K, Ohsugi M, Shirahige K. Inferring the choreography of parental genomes during fertilization from ultralarge-scale whole-transcriptome analysis. Genes Dev 2014; 27:2736-48. [PMID: 24352427 PMCID: PMC3877761 DOI: 10.1101/gad.227926.113] [Citation(s) in RCA: 81] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
The mechanisms underlying parental genome coordination upon mammalian fertilization remain elusive due to difficulties in preparing large numbers of high-quality preimplantation cells. Here, Park et al. collect an unprecedented number of mouse oocytes and establish detailed transcriptional profiles for four early embryonic stages and parthenogenetic development. Bioinformatic analysis identifies a distinctive gene regulatory network activated in embryos after fertilization compared with parthenotes. This large-scale profile of early mouse embryos yields a valuable resource for developmental biology and stem cell research. Fertilization precisely choreographs parental genomes by using gamete-derived cellular factors and activating genome regulatory programs. However, the mechanism remains elusive owing to the technical difficulties of preparing large numbers of high-quality preimplantation cells. Here, we collected >14 × 104 high-quality mouse metaphase II oocytes and used these to establish detailed transcriptional profiles for four early embryo stages and parthenogenetic development. By combining these profiles with other public resources, we found evidence that gene silencing appeared to be mediated in part by noncoding RNAs and that this was a prerequisite for post-fertilization development. Notably, we identified 817 genes that were differentially expressed in embryos after fertilization compared with parthenotes. The regulation of these genes was distinctly different from those expressed in parthenotes, suggesting functional specialization of particular transcription factors prior to first cell cleavage. We identified five transcription factors that were potentially necessary for developmental progression: Foxd1, Nkx2-5, Sox18, Myod1, and Runx1. Our very large-scale whole-transcriptome profile of early mouse embryos yielded a novel and valuable resource for studies in developmental biology and stem cell research. The database is available at http://dbtmee.hgc.jp.
Collapse
Affiliation(s)
- Sung-Joon Park
- Human Genome Center, the Institute of Medical Science, the University of Tokyo, Tokyo 108-8639, Japan
| | | | | | | | | | | | | |
Collapse
|
190
|
Pei Z, Chen X, Sun C, Du H, Wei H, Song W, Yang Y, Zhang M, Lu W, Cheng R, Luo F. A novel single nucleotide polymorphism in the protein tyrosine phosphatase N22 gene (PTPN22) is associated with Type 1 diabetes in a Chinese population. Diabet Med 2014; 31:219-26. [PMID: 24117662 DOI: 10.1111/dme.12331] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2013] [Revised: 08/09/2013] [Accepted: 09/19/2013] [Indexed: 11/29/2022]
Abstract
AIMS To examine single nucleotide polymorphisms in the protein tyrosine phosphatase N22 gene (PTPN22) and to study their association with Type 1 diabetes in a Chinese cohort. METHODS Three hundred and sixty-four young patients with Type 1 diabetes and 719 healthy children were included in this case-controlled study. The genotypes of rs1217385, rs2488457 (-1123C>G), rs1217414, rs1217419, rs3765598 and rs2476601 (1858C>T) in the PTPN22 gene were determined using the SNaPshot method. Alleles, genotypes and haplotype frequencies were compared between patients with Type 1 diabetes and healthy control subjects. The association between single nucleotide polymorphisms and clinical traits/autoantibody status was also analysed. RESULTS The single nucleotide polymorphism, rs1217419, located in the second intron of the PTPN22 gene was associated with Type 1 diabetes (odds ratio 1.5, 95% CI 1.14-1.97, P = 0.003). An additional single nucleotide polymorphism, rs1217385, was also associated with Type 1 diabetes; however, the association was secondary to that of rs1217419. The previously reported single nucleotide polymorphism that is associated with Type 1 diabetes (-1123G>C) had only marginal association with Type 1 diabetes in our study. A marginal association was also identified between -1123G>C and glutamic acid decarboxylase autoantibody positivity in patients with Type 1 diabetes. There was no association between the single nucleotide polymorphism 1858C>T and Type 1 diabetes in our studied cohort. CONCLUSIONS Our study confirmed that PTPN22 is a gene that contributes to Type 1 diabetes susceptibility. The primary association occurs with single nucleotide polymorphism rs1217419 and there is clear heterogeneity of the association between PTPTN22 polymorphisms and Type 1 diabetes in a Chinese population compared with other populations.
Collapse
Affiliation(s)
- Z Pei
- Department of Pediatric Endocrinology and Inborn Metabolic Diseases, Children's Hospital of Fudan University, Shanghai, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
191
|
Vonlanthen J, Okoniewski MJ, Menigatti M, Cattaneo E, Pellegrini-Ochsner D, Haider R, Jiricny J, Staiano T, Buffoli F, Marra G. A comprehensive look at transcription factor gene expression changes in colorectal adenomas. BMC Cancer 2014; 14:46. [PMID: 24472434 PMCID: PMC4078005 DOI: 10.1186/1471-2407-14-46] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2013] [Accepted: 01/23/2014] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Biological processes are controlled by transcription networks. Expression changes of transcription factor (TF) genes in precancerous lesions are therefore crucial events in tumorigenesis. Our aim was to obtain a comprehensive picture of these changes in colorectal adenomas. METHODS Using a 3-pronged selection procedure, we analyzed transcriptomic data on 34 human tissue samples (17 adenomas and paired samples of normal mucosa, all collected with ethics committee approval and written, informed patient consent) to identify TFs with highly significant tumor-associated gene expression changes whose potential roles in colorectal tumorigenesis have been under-researched. Microarray data were subjected to stringent statistical analysis of TF expression in tumor vs. normal tissues, MetaCore-mediated identification of TF networks displaying enrichment for genes that were differentially expressed in tumors, and a novel quantitative analysis of the publications examining the TF genes' roles in colorectal tumorigenesis. RESULTS The 261 TF genes identified with this procedure included DACH1, which plays essential roles in the proper proliferation and differentiation of retinal and leg precursor cell populations in Drosophila melanogaster. Its possible roles in colorectal tumorigenesis are completely unknown, but it was found to be markedly overexpressed (mRNA and protein) in all colorectal adenomas and in most colorectal carcinomas. However, DACH1 expression was absent in some carcinomas, most of which were DNA mismatch-repair deficient. When networks were built using the set of TF genes identified by all three selection procedures, as well as the entire set of transcriptomic changes in adenomas, five hub genes (TGFB1, BIRC5, MYB, NR3C1, and TERT) where identified as putatively crucial components of the adenomatous transformation process. CONCLUSION The transcription-regulating network of colorectal adenomas (compared with that of normal colorectal mucosa) is characterized by significantly altered expression of over 250 TF genes, many of which have never been investigated in relation to colorectal tumorigenesis.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Giancarlo Marra
- Institute of Molecular Cancer Research, University of Zurich, Winterthurerstrasse 190, Zurich 8051, Switzerland.
| |
Collapse
|
192
|
Chen H, VanBuren V. A provisional gene regulatory atlas for mouse heart development. PLoS One 2014; 9:e83364. [PMID: 24421884 PMCID: PMC3885437 DOI: 10.1371/journal.pone.0083364] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2013] [Accepted: 11/02/2013] [Indexed: 12/29/2022] Open
Abstract
Congenital Heart Disease (CHD) is one of the most common birth defects. Elucidating the molecular mechanisms underlying normal cardiac development is an important step towards early identification of abnormalities during the developmental program and towards the creation of early intervention strategies. We developed a novel computational strategy for leveraging high-content data sets, including a large selection of microarray data associated with mouse cardiac development, mouse genome sequence, ChIP-seq data of selected mouse transcription factors and Y2H data of mouse protein-protein interactions, to infer the active transcriptional regulatory network of mouse cardiac development. We identified phase-specific expression activity for 765 overlapping gene co-expression modules that were defined for obtained cardiac lineage microarray data. For each co-expression module, we identified the phase of cardiac development where gene expression for that module was higher than other phases. Co-expression modules were found to be consistent with biological pathway knowledge in Wikipathways, and met expectations for enrichment of pathways involved in heart lineage development. Over 359,000 transcription factor-target relationships were inferred by analyzing the promoter sequences within each gene module for overrepresentation against the JASPAR database of Transcription Factor Binding Site (TFBS) motifs. The provisional regulatory network will provide a framework of studying the genetic basis of CHD.
Collapse
Affiliation(s)
- Hailin Chen
- Department of Medical Physiology, Texas A&M HSC College of Medicine, Temple, Texas, United States of America
| | - Vincent VanBuren
- Department of Medical Physiology, Texas A&M HSC College of Medicine, Temple, Texas, United States of America
- * E-mail:
| |
Collapse
|
193
|
Le HS, Bar-Joseph Z. Integrating sequence, expression and interaction data to determine condition-specific miRNA regulation. Bioinformatics 2013; 29:i89-97. [PMID: 23813013 PMCID: PMC3694655 DOI: 10.1093/bioinformatics/btt231] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Motivation: MicroRNAs (miRNAs) are small non-coding RNAs that regulate gene expression post-transcriptionally. MiRNAs were shown to play an important role in development and disease, and accurately determining the networks regulated by these miRNAs in a specific condition is of great interest. Early work on miRNA target prediction has focused on using static sequence information. More recently, researchers have combined sequence and expression data to identify such targets in various conditions. Results: We developed the Protein Interaction-based MicroRNA Modules (PIMiM), a regression-based probabilistic method that integrates sequence, expression and interaction data to identify modules of mRNAs controlled by small sets of miRNAs. We formulate an optimization problem and develop a learning framework to determine the module regulation and membership. Applying PIMiM to cancer data, we show that by adding protein interaction data and modeling cooperative regulation of mRNAs by a small number of miRNAs, PIMiM can accurately identify both miRNA and their targets improving on previous methods. We next used PIMiM to jointly analyze a number of different types of cancers and identified both common and cancer-type-specific miRNA regulators. Contact:zivbj@cs.cmu.edu Supplementary information:Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Hai-Son Le
- Machine Learning Department, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | | |
Collapse
|
194
|
Zhang W, Zhao J, Lee JF, Gartung A, Jawadi H, Lambiv WL, Honn KV, Lee MJ. ETS-1-mediated transcriptional up-regulation of CD44 is required for sphingosine-1-phosphate receptor subtype 3-stimulated chemotaxis. J Biol Chem 2013; 288:32126-32137. [PMID: 24064218 PMCID: PMC3820853 DOI: 10.1074/jbc.m113.495218] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2013] [Revised: 09/18/2013] [Indexed: 12/17/2022] Open
Abstract
Sphingosine-1-phosphate (S1P)-regulated chemotaxis plays critical roles in various physiological and pathophysiological conditions. S1P-regulated chemotaxis is mediated by the S1P family of G-protein-coupled receptors. However, molecular details of the S1P-regulated chemotaxis are incompletely understood. Cultured human lung adenocarcinoma cell lines abundantly express S1P receptor subtype 3 (S1P3), thus providing a tractable in vitro system to characterize molecular mechanism(s) underlying the S1P3 receptor-regulated chemotactic response. S1P treatment enhances CD44 expression and induces membrane localization of CD44 polypeptides via the S1P3/Rho kinase (ROCK) signaling pathway. Knockdown of CD44 completely diminishes the S1P-stimulated chemotaxis. Promoter analysis suggests that the CD44 promoter contains binding sites of the ETS-1 (v-ets erythroblastosis virus E26 oncogene homolog 1) transcriptional factor. ChIP assay confirms that S1P treatment stimulates the binding of ETS-1 to the CD44 promoter region. Moreover, S1P induces the expression and nuclear translocation of ETS-1. Knockdown of S1P3 or inhibition of ROCK abrogates the S1P-induced ETS-1 expression. Furthermore, knockdown of ETS-1 inhibits the S1P-induced CD44 expression and cell migration. In addition, we showed that S1P3/ROCK signaling up-regulates ETS-1 via the activity of JNK. Collectively, we characterized a novel signaling axis, i.e., ROCK-JNK-ETS-1-CD44 pathway, which plays an essential role in the S1P3-regulated chemotactic response.
Collapse
Affiliation(s)
- Wenliang Zhang
- From the Department of Pathology,; the Bioactive Lipid Research Program
| | - Jiawei Zhao
- From the Department of Pathology,; the Bioactive Lipid Research Program
| | - Jen-Fu Lee
- From the Department of Pathology,; the Bioactive Lipid Research Program
| | - Allison Gartung
- From the Department of Pathology,; the Bioactive Lipid Research Program
| | | | | | - Kenneth V Honn
- From the Department of Pathology,; the Bioactive Lipid Research Program,; the Karmanos Cancer Institute
| | - Menq-Jer Lee
- From the Department of Pathology,; the Bioactive Lipid Research Program,; the Karmanos Cancer Institute; the Cardiovascular Research Institute, Wayne State University School of Medicine, Detroit, Michigan 48201.
| |
Collapse
|
195
|
Noh JY, Patnaik BB, Tindwa H, Seo GW, Kim DH, Patnaik HH, Jo YH, Lee YS, Lee BL, Kim NJ, Han YS. Genomic organization, sequence characterization and expression analysis of Tenebrio molitor apolipophorin-III in response to an intracellular pathogen, Listeria monocytogenes. Gene 2013; 534:204-17. [PMID: 24200961 DOI: 10.1016/j.gene.2013.10.058] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2013] [Revised: 10/21/2013] [Accepted: 10/26/2013] [Indexed: 11/29/2022]
Abstract
Apolipophorin III (apoLp-III) is a well-known hemolymph protein having a functional role in lipid transport and immune response of insects. We cloned full-length cDNA encoding putative apoLp-III from larvae of the coleopteran beetle, Tenebrio molitor (TmapoLp-III), by identification of clones corresponding to the partial sequence of TmapoLp-III, subsequently followed with full length sequencing by a clone-by-clone primer walking method. The complete cDNA consists of 890 nucleotides, including an ORF encoding 196 amino acid residues. Excluding a putative signal peptide of the first 20 amino acid residues, the 176-residue mature apoLp-III has a calculated molecular mass of 19,146Da. Genomic sequence analysis with respect to its cDNA showed that TmapoLp-III was organized into four exons interrupted by three introns. Several immune-related transcription factor binding sites were discovered in the putative 5'-flanking region. BLAST and phylogenetic analyses reveal that TmapoLp-III has high sequence identity (88%) with Tribolium castaneum apoLp-III but shares little sequence homologies (<26%) with other apoLp-IIIs. Homology modeling of Tm apoLp-III shows a bundle of five amphipathic alpha helices, including a short helix 3'. The 'helix-short helix-helix' motif was predicted to be implicated in lipid binding interactions, through reversible conformational changes and accommodating the hydrophobic residues to the exterior for stability. Highest level of TmapoLp-III mRNA was detected at late pupal stages, albeit it is expressed in the larval and adult stages at lower levels. The tissue specific expression of the transcripts showed significantly higher numbers in larval fat body and adult integument. In addition, TmapoLp-III mRNA was found to be highly upregulated in late stages of L. monocytogenes or E. coli challenge. These results indicate that TmapoLp-III may play an important role in innate immune responses against bacterial pathogens in T. molitor.
Collapse
Affiliation(s)
- Ju Young Noh
- Division of Plant Biotechnology, Institute of Environmentally-Friendly Agriculture (IEFA), College of Agriculture and Life Sciences, Chonnam National University, Gwangju 500-757, Republic of Korea
| | - Bharat Bhusan Patnaik
- Division of Plant Biotechnology, Institute of Environmentally-Friendly Agriculture (IEFA), College of Agriculture and Life Sciences, Chonnam National University, Gwangju 500-757, Republic of Korea
| | - Hamisi Tindwa
- Division of Plant Biotechnology, Institute of Environmentally-Friendly Agriculture (IEFA), College of Agriculture and Life Sciences, Chonnam National University, Gwangju 500-757, Republic of Korea
| | - Gi Won Seo
- Division of Plant Biotechnology, Institute of Environmentally-Friendly Agriculture (IEFA), College of Agriculture and Life Sciences, Chonnam National University, Gwangju 500-757, Republic of Korea
| | - Dong Hyun Kim
- Division of Plant Biotechnology, Institute of Environmentally-Friendly Agriculture (IEFA), College of Agriculture and Life Sciences, Chonnam National University, Gwangju 500-757, Republic of Korea
| | - Hongray Howrelia Patnaik
- Division of Plant Biotechnology, Institute of Environmentally-Friendly Agriculture (IEFA), College of Agriculture and Life Sciences, Chonnam National University, Gwangju 500-757, Republic of Korea
| | - Yong Hun Jo
- Division of Plant Biotechnology, Institute of Environmentally-Friendly Agriculture (IEFA), College of Agriculture and Life Sciences, Chonnam National University, Gwangju 500-757, Republic of Korea
| | - Yong Seok Lee
- Department of Life Science and Biotechnology, College of Natural Sciences, Soonchunhyang University, Asan City 336-745 Republic of Korea
| | - Bok Luel Lee
- National Research Laboratory of Defense Proteins, College of Pharmacy, Pusan National University, Jangjeon Dong, Kumjeong Ku, Busan, 609-735, Republic of Korea
| | - Nam Jung Kim
- Division of Applied Entomology, National Academy of Agricultural Science, Rural Development, 61th, Seodun-dong, Gwonseon-gu, Suwon, Gyeonggi-do, 441-853, Republic of Korea
| | - Yeon Soo Han
- Division of Plant Biotechnology, Institute of Environmentally-Friendly Agriculture (IEFA), College of Agriculture and Life Sciences, Chonnam National University, Gwangju 500-757, Republic of Korea.
| |
Collapse
|
196
|
Chen Z, Shentu TP, Wen L, Johnson DA, Shyy JYJ. Regulation of SIRT1 by oxidative stress-responsive miRNAs and a systematic approach to identify its role in the endothelium. Antioxid Redox Signal 2013; 19:1522-38. [PMID: 23477488 PMCID: PMC3797452 DOI: 10.1089/ars.2012.4803] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
SIGNIFICANCE Oxidative stress is a common denominator of various risk factors contributing to endothelial dysfunction and vascular diseases. Accumulated evidence suggests that sirtuin 1 (SIRT1) expression and/or activity is impaired by supraphysiological levels of oxidative stress, which in turn disrupts endothelial homeostasis. RECENT ADVANCES Several microRNAs (miRNAs) are induced by oxidative stress and termed as oxidative stress-responsive miRNAs. They may play a role linking the imbalanced redox state with dysregulated SIRT1. CRITICAL ISSUES This review summarizes recent findings on oxidative stress-responsive miRNAs and their involvement in SIRT1 regulation. Because of the unique characteristics of miRNAs, research in this new area requires an integrative approach that combines bioinformatics and experimental validation. Thus, a research strategy is discussed to identify the SIRT1-regulating miRNAs under oxidative stress and their functional outcomes in relation to endothelial dysfunction. Additionally, the miRNAs implicated in vascular diseases such as atherosclerosis and abdominal aortic aneurysms are discussed along with the translational potential and challenges of using miRNAs and its analogs as therapeutic agents. FUTURE DIRECTIONS Although at its infancy, research on oxidative stress-responsive miRNAs and their regulation of SIRT1 may provide new insights in understanding vascular disorders. Moreover, systematic approaches integrating in silico, in vitro, and in vivo observations can be useful tools in revealing the pathways modulating endothelial biology.
Collapse
Affiliation(s)
- Zhen Chen
- 1 Division of Biomedical Sciences, University of California , Riverside, California
| | | | | | | | | |
Collapse
|
197
|
Lane A, Segura-Cabrera A, Komurov K. A comparative survey of functional footprints of EGFR pathway mutations in human cancers. Oncogene 2013; 33:5078-89. [PMID: 24166508 DOI: 10.1038/onc.2013.452] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2013] [Revised: 09/13/2013] [Accepted: 09/20/2013] [Indexed: 12/19/2022]
Abstract
Genes functioning in epidermal growth factor receptor (EGFR) signaling pathways are among the most frequently activated oncogenes in human cancers. We have conducted a comparative analysis of functional footprints (that is, effect on signaling and transcriptional landscapes in cells) associated with oncogenic and tumor suppressor mutations in EGFR pathway genes in human cancers. We have found that mutations in the EGFR pathway differentially have an impact on signaling and metabolic pathways in cancer cells in a mutation- and tissue-selective manner. For example, although signaling and metabolic profiles of breast tumors with PIK3CA or AKT1 mutations are, as expected, highly similar, they display markedly different, sometimes even opposite, profiles to those with ERBB2 or EGFR amplifications. On the other hand, although low-grade gliomas and glioblastomas, both brain cancers, driven by EGFR amplifications are highly functionally similar, their functional footprints are significantly different from lung and breast tumors driven by EGFR or ERBB2. Overall, these observations argue that, contrary to expectations, the mechanisms of tumorigenicity associated with mutations in different genes along the same pathway, or in the same gene across different tissues, may be highly different. We present evidence that oncogenic functional footprints in cancer cell lines have significantly diverged from those in tumor tissues, which potentially explains the discrepancy of our findings with the current knowledge. Nevertheless, our analyses reveal a common inflammatory response signature in EGFR-driven human cancers of different tissue origins. Our results may have implications in the design of therapeutic strategies in cancers driven by these oncogenes.
Collapse
Affiliation(s)
- A Lane
- Division of Experimental Hematology and Cancer Biology, Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - A Segura-Cabrera
- Division of Experimental Hematology and Cancer Biology, Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - K Komurov
- 1] Division of Experimental Hematology and Cancer Biology, Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA [2] Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA [3] Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| |
Collapse
|
198
|
Brand LH, Henneges C, Schüssler A, Kolukisaoglu HÜ, Koch G, Wallmeroth N, Hecker A, Thurow K, Zell A, Harter K, Wanke D. Screening for protein-DNA interactions by automatable DNA-protein interaction ELISA. PLoS One 2013; 8:e75177. [PMID: 24146751 PMCID: PMC3795721 DOI: 10.1371/journal.pone.0075177] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2013] [Accepted: 08/12/2013] [Indexed: 12/22/2022] Open
Abstract
DNA-binding proteins (DBPs), such as transcription factors, constitute about 10% of the protein-coding genes in eukaryotic genomes and play pivotal roles in the regulation of chromatin structure and gene expression by binding to short stretches of DNA. Despite their number and importance, only for a minor portion of DBPs the binding sequence had been disclosed. Methods that allow the de novo identification of DNA-binding motifs of known DBPs, such as protein binding microarray technology or SELEX, are not yet suited for high-throughput and automation. To close this gap, we report an automatable DNA-protein-interaction (DPI)-ELISA screen of an optimized double-stranded DNA (dsDNA) probe library that allows the high-throughput identification of hexanucleotide DNA-binding motifs. In contrast to other methods, this DPI-ELISA screen can be performed manually or with standard laboratory automation. Furthermore, output evaluation does not require extensive computational analysis to derive a binding consensus. We could show that the DPI-ELISA screen disclosed the full spectrum of binding preferences for a given DBP. As an example, AtWRKY11 was used to demonstrate that the automated DPI-ELISA screen revealed the entire range of in vitro binding preferences. In addition, protein extracts of AtbZIP63 and the DNA-binding domain of AtWRKY33 were analyzed, which led to a refinement of their known DNA-binding consensi. Finally, we performed a DPI-ELISA screen to disclose the DNA-binding consensus of a yet uncharacterized putative DBP, AtTIFY1. A palindromic TGATCA-consensus was uncovered and we could show that the GATC-core is compulsory for AtTIFY1 binding. This specific interaction between AtTIFY1 and its DNA-binding motif was confirmed by in vivo plant one-hybrid assays in protoplasts. Thus, the value and applicability of the DPI-ELISA screen for de novo binding site identification of DBPs, also under automatized conditions, is a promising approach for a deeper understanding of gene regulation in any organism of choice.
Collapse
Affiliation(s)
- Luise H. Brand
- Plant Physiology, Center for Plant Molecular Biology, University of Tuebingen, Tuebingen, Germany
| | - Carsten Henneges
- Cognitive Systems, Center for Bioinformatics, University of Tuebingen, Tuebingen, Germany
| | - Axel Schüssler
- Cognitive Systems, Center for Bioinformatics, University of Tuebingen, Tuebingen, Germany
| | - H. Üner Kolukisaoglu
- Plant Physiology, Center for Plant Molecular Biology, University of Tuebingen, Tuebingen, Germany
- Center for Life Science Automation, Rostock, Germany
| | - Grit Koch
- Center for Life Science Automation, Rostock, Germany
| | - Niklas Wallmeroth
- Plant Physiology, Center for Plant Molecular Biology, University of Tuebingen, Tuebingen, Germany
| | - Andreas Hecker
- Plant Physiology, Center for Plant Molecular Biology, University of Tuebingen, Tuebingen, Germany
| | | | - Andreas Zell
- Cognitive Systems, Center for Bioinformatics, University of Tuebingen, Tuebingen, Germany
| | - Klaus Harter
- Plant Physiology, Center for Plant Molecular Biology, University of Tuebingen, Tuebingen, Germany
| | - Dierk Wanke
- Plant Physiology, Center for Plant Molecular Biology, University of Tuebingen, Tuebingen, Germany
- * E-mail:
| |
Collapse
|
199
|
Jiang W, Zhang Y, Meng F, Lian B, Chen X, Yu X, Dai E, Wang S, Liu X, Li X, Wang L, Li X. Identification of active transcription factor and miRNA regulatory pathways in Alzheimer’s disease. Bioinformatics 2013; 29:2596-602. [DOI: 10.1093/bioinformatics/btt423] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
200
|
Soares MPM, Barchuk AR, Simões ACQ, Dos Santos Cristino A, de Paula Freitas FC, Canhos LL, Bitondi MMG. Genes involved in thoracic exoskeleton formation during the pupal-to-adult molt in a social insect model, Apis mellifera. BMC Genomics 2013; 14:576. [PMID: 23981317 PMCID: PMC3766229 DOI: 10.1186/1471-2164-14-576] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2013] [Accepted: 08/23/2013] [Indexed: 12/04/2022] Open
Abstract
Background The insect exoskeleton provides shape, waterproofing, and locomotion via attached somatic muscles. The exoskeleton is renewed during molting, a process regulated by ecdysteroid hormones. The holometabolous pupa transforms into an adult during the imaginal molt, when the epidermis synthe3sizes the definitive exoskeleton that then differentiates progressively. An important issue in insect development concerns how the exoskeletal regions are constructed to provide their morphological, physiological and mechanical functions. We used whole-genome oligonucleotide microarrays to screen for genes involved in exoskeletal formation in the honeybee thoracic dorsum. Our analysis included three sampling times during the pupal-to-adult molt, i.e., before, during and after the ecdysteroid-induced apolysis that triggers synthesis of the adult exoskeleton. Results Gene ontology annotation based on orthologous relationships with Drosophila melanogaster genes placed the honeybee differentially expressed genes (DEGs) into distinct categories of Biological Process and Molecular Function, depending on developmental time, revealing the functional elements required for adult exoskeleton formation. Of the 1,253 unique DEGs, 547 were upregulated in the thoracic dorsum after apolysis, suggesting induction by the ecdysteroid pulse. The upregulated gene set included 20 of the 47 cuticular protein (CP) genes that were previously identified in the honeybee genome, and three novel putative CP genes that do not belong to a known CP family. In situ hybridization showed that two of the novel genes were abundantly expressed in the epidermis during adult exoskeleton formation, strongly implicating them as genuine CP genes. Conserved sequence motifs identified the CP genes as members of the CPR, Tweedle, Apidermin, CPF, CPLCP1 and Analogous-to-Peritrophins families. Furthermore, 28 of the 36 muscle-related DEGs were upregulated during the de novo formation of striated fibers attached to the exoskeleton. A search for cis-regulatory motifs in the 5′-untranslated region of the DEGs revealed potential binding sites for known transcription factors. Construction of a regulatory network showed that various upregulated CP- and muscle-related genes (15 and 21 genes, respectively) share common elements, suggesting co-regulation during thoracic exoskeleton formation. Conclusions These findings help reveal molecular aspects of rigid thoracic exoskeleton formation during the ecdysteroid-coordinated pupal-to-adult molt in the honeybee.
Collapse
Affiliation(s)
- Michelle Prioli Miranda Soares
- Departamento de Biologia, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, Brasil.
| | | | | | | | | | | | | |
Collapse
|