51
|
A reference set of curated biomedical data and metadata from clinical case reports. Sci Data 2018; 5:180258. [PMID: 30457569 PMCID: PMC6244181 DOI: 10.1038/sdata.2018.258] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Accepted: 09/27/2018] [Indexed: 12/30/2022] Open
Abstract
Clinical case reports (CCRs) provide an important means of sharing clinical experiences about atypical disease phenotypes and new therapies. However, published case reports contain largely unstructured and heterogeneous clinical data, posing a challenge to mining relevant information. Current indexing approaches generally concern document-level features and have not been specifically designed for CCRs. To address this disparity, we developed a standardized metadata template and identified text corresponding to medical concepts within 3,100 curated CCRs spanning 15 disease groups and more than 750 reports of rare diseases. We also prepared a subset of metadata on reports on selected mitochondrial diseases and assigned ICD-10 diagnostic codes to each. The resulting resource, Metadata Acquired from Clinical Case Reports (MACCRs), contains text associated with high-level clinical concepts, including demographics, disease presentation, treatments, and outcomes for each report. Our template and MACCR set render CCRs more findable, accessible, interoperable, and reusable (FAIR) while serving as valuable resources for key user groups, including researchers, physician investigators, clinicians, data scientists, and those shaping government policies for clinical trials.
Collapse
|
52
|
Leal L, Guardo AC, Morón-López S, Salgado M, Mothe B, Heirman C, Pannus P, Vanham G, van den Ham HJ, Gruters R, Andeweg A, Van Meirvenne S, Pich J, Arnaiz JA, Gatell JM, Brander C, Thielemans K, Martínez-Picado J, Plana M, García F. Phase I clinical trial of an intranodally administered mRNA-based therapeutic vaccine against HIV-1 infection. AIDS 2018; 32:2533-2545. [PMID: 30289805 PMCID: PMC6221380 DOI: 10.1097/qad.0000000000002026] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Objective: The efficacy of therapeutic vaccines against HIV-1 infection has been modest. New inerts to redirect responses to vulnerable sites are urgently needed to improve these results. Design: We performed the first-in-human clinical trial with naked mRNA (iHIVARNA) combining a dendritic cell activation strategy (TriMix:CD40L+CD70+caTLR4 RNA) with a novel HIV immunogen sequences (HTI immunogen). Methods: A dose escalation, phase I clinical trial was performed in 21 chronic HIV-1-infected patients under ART who received three intranodal doses of mRNA (weeks 0, 2 and 4) as follow: TriMix-100 g, TriMix-300 g, TriMix-300 g with HTI-300 g, TriMix-300 g with HTI-600 g, TriMix-300 g with HTI-900 g. Primary end-point was safety and secondary-exploratory end-points were immunogenicity, changes in viral reservoir and transcriptome. Results: Overall, the vaccine was secure and well tolerated. There were 31 grade 1/2 and 1 grade 3 adverse events, mostly unrelated to the vaccination. Patients who received the highest dose showed a moderate increase in T-cell responses spanning HTI sequence at week 8. In addition, the proportion of responders receiving any dose of HTI increased from 31% at w0 to 80% postvaccination. The intervention had no impact on caHIV-DNA levels, however, caHIV-RNA expression and usVL were transiently increased at weeks 5 and 6 in the highest dose of iHIVARNA, and these changes were positively correlated with HIV-1-specific-induced immune responses. Conclusion: This phase I dose-escalating trial showed that iHIVARNA administration was safe and well tolerated, induced moderate HIV-specific T-cell responses and transiently increased different viral replication readouts. These data support further exploration of iHIVARNA in a phase II study. ClinicalTrials.gov Identifier: NCT02413645
Collapse
|
53
|
Ling A, Gruener RF, Fessler J, Huang RS. More than fishing for a cure: The promises and pitfalls of high throughput cancer cell line screens. Pharmacol Ther 2018; 191:178-189. [PMID: 29953899 PMCID: PMC7001883 DOI: 10.1016/j.pharmthera.2018.06.014] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
High-throughput screens in cancer cell lines (CCLs) have been used for decades to help researchers identify compounds with the potential to improve the treatment of cancer and, more recently, to identify genomic susceptibilities in cancer via genome-wide shRNA and CRISPR/Cas9 screens. Additionally, rich genomic and transcriptomic data of these CCLs has allowed researchers to pair this screening data with biological features, enabling efforts to identify biomarkers of treatment response and gene dependencies. In this paper, we review the major CCL screening efforts and the large datasets these screens have made available. We also assess the CCL screens collectively and include a resource with harmonized CCL and compound identifiers to facilitate comparisons across screens. The CCLs in these screens were found to represent a wide range of cancer types, with a strong correlation between the representation of a cancer type and its associated mortality. Patient ages and gender distributions of CCLs were generally as expected, with some notable exceptions of female underrepresentation in certain disease types. Also, ethnicity information, while largely incomplete, suggests that African American and Hispanic patients may be severely underrepresented in these screens. Nearly all genes were targeted in the genetic perturbations screens, but the compounds used for the drug screens target less than half of known cancer drivers, likely reflecting known limitations in our drug design capabilities. Finally, we discuss recent developments in the field and the promise they hold for enabling future screens to overcome previous limitations and lead to new breakthroughs in cancer treatment.
Collapse
Affiliation(s)
- Alexander Ling
- Committee on Cancer Biology, University of Chicago, Chicago, IL, United States; Department of Experimental and Clinical Pharmacology, University of Minnesota, Minneapolis, MN, United States
| | - Robert F Gruener
- Committee on Cancer Biology, University of Chicago, Chicago, IL, United States; Ben May Department for Cancer Research, University of Chicago, Chicago, IL, United States
| | - Jessica Fessler
- Committee on Cancer Biology, University of Chicago, Chicago, IL, United States; Department of Pathology, University of Chicago, Chicago, IL, United States
| | - R Stephanie Huang
- Department of Experimental and Clinical Pharmacology, University of Minnesota, Minneapolis, MN, United States.
| |
Collapse
|
54
|
Burger B, Hernández Sánchez LF, Lereim RR, Barsnes H, Vaudel M. Analyzing the Structure of Pathways and Its Influence on the Interpretation of Biomedical Proteomics Data Sets. J Proteome Res 2018; 17:3801-3809. [PMID: 30251541 DOI: 10.1021/acs.jproteome.8b00464] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Biochemical pathways are commonly used as a reference to conduct functional analysis on biomedical omics data sets, where experimental results are mapped to knowledgebases comprising known molecular interactions collected from the literature. Due to their central role, the content of the functional knowledgebases directly influences the outcome of pathway analyses. In this study, we investigate the structure of the current pathway knowledge, as exemplified by Reactome, discuss the consequences for biological interpretation, and outline possible improvements in the use of pathway knowledgebases. By providing a view of the underlying protein interaction network, we aim to help pathway analysis users manage their expectations and better identify possible artifacts in the results.
Collapse
Affiliation(s)
- Bram Burger
- Computational Biology Unit (CBU), Department of Informatics , University of Bergen , 5020 Bergen , Norway.,Proteomics Unit (PROBE), Department of Biomedicine , University of Bergen , 5020 Bergen , Norway
| | - Luis Francisco Hernández Sánchez
- KG Jebsen Center for Diabetes Research, Department of Clinical Science , University of Bergen , 5020 Bergen , Norway.,Center for Medical Genetics and Molecular Medicine , Haukeland University Hospital , 5020 Bergen , Norway
| | - Ragnhild Reehorst Lereim
- Computational Biology Unit (CBU), Department of Informatics , University of Bergen , 5020 Bergen , Norway.,Proteomics Unit (PROBE), Department of Biomedicine , University of Bergen , 5020 Bergen , Norway
| | - Harald Barsnes
- Computational Biology Unit (CBU), Department of Informatics , University of Bergen , 5020 Bergen , Norway.,Proteomics Unit (PROBE), Department of Biomedicine , University of Bergen , 5020 Bergen , Norway
| | - Marc Vaudel
- KG Jebsen Center for Diabetes Research, Department of Clinical Science , University of Bergen , 5020 Bergen , Norway.,Center for Medical Genetics and Molecular Medicine , Haukeland University Hospital , 5020 Bergen , Norway
| |
Collapse
|
55
|
Kishinevsky S, Wang T, Rodina A, Chung SY, Xu C, Philip J, Taldone T, Joshi S, Alpaugh ML, Bolaender A, Gutbier S, Sandhu D, Fattahi F, Zimmer B, Shah SK, Chang E, Inda C, Koren J, Saurat NG, Leist M, Gross SS, Seshan VE, Klein C, Tomishima MJ, Erdjument-Bromage H, Neubert TA, Henrickson RC, Chiosis G, Studer L. HSP90-incorporating chaperome networks as biosensor for disease-related pathways in patient-specific midbrain dopamine neurons. Nat Commun 2018; 9:4345. [PMID: 30341316 PMCID: PMC6195591 DOI: 10.1038/s41467-018-06486-6] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Accepted: 08/29/2018] [Indexed: 12/21/2022] Open
Abstract
Environmental and genetic risk factors contribute to Parkinson’s Disease (PD) pathogenesis and the associated midbrain dopamine (mDA) neuron loss. Here, we identify early PD pathogenic events by developing methodology that utilizes recent innovations in human pluripotent stem cells (hPSC) and chemical sensors of HSP90-incorporating chaperome networks. We show that events triggered by PD-related genetic or toxic stimuli alter the neuronal proteome, thereby altering the stress-specific chaperome networks, which produce changes detected by chemical sensors. Through this method we identify STAT3 and NF-κB signaling activation as examples of genetic stress, and phospho-tyrosine hydroxylase (TH) activation as an example of toxic stress-induced pathways in PD neurons. Importantly, pharmacological inhibition of the stress chaperome network reversed abnormal phospho-STAT3 signaling and phospho-TH-related dopamine levels and rescued PD neuron viability. The use of chemical sensors of chaperome networks on hPSC-derived lineages may present a general strategy to identify molecular events associated with neurodegenerative diseases. The early molecular events that ultimately lead to neuronal cell death in pathologies such as Parkinson’s disease are poorly understood. Here the authors use pluripotent stem-cell-derived human midbrain neurons and chemical biology tools to gain molecular level insight into the events induced by toxic and genetic stresses that mimic those occurring during neurodegeneration.
Collapse
Affiliation(s)
- Sarah Kishinevsky
- The Center for Stem Cell Biology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, Box 256, New York, NY, 10065, USA.,Developmental Biology Program, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, Box 256, New York, NY, 10065, USA.,Program in Chemical Biology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA.,Neuroscience Graduate Program of Weill Cornell Graduate School of Biomedical Sciences, Weill Cornell Medical College, 1300 York Avenue, Box 65, New York, NY, 10065, USA
| | - Tai Wang
- Program in Chemical Biology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| | - Anna Rodina
- Program in Chemical Biology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| | - Sun Young Chung
- The Center for Stem Cell Biology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, Box 256, New York, NY, 10065, USA.,Developmental Biology Program, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, Box 256, New York, NY, 10065, USA
| | - Chao Xu
- Program in Chemical Biology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| | - John Philip
- Proteomics Core Facility, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| | - Tony Taldone
- Program in Chemical Biology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| | - Suhasini Joshi
- Program in Chemical Biology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| | - Mary L Alpaugh
- Program in Chemical Biology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA.,Department of Molecular and Cellular Biosciences, Rowan University, 1275 York Avenue, Glassboro, NJ, 08028, USA
| | - Alexander Bolaender
- Program in Chemical Biology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| | - Simon Gutbier
- Doerenkamp-Zbinden Chair for In Vitro Toxicology and Biomedicine, University of Konstanz, Konstanz, 78464, Germany
| | - Davinder Sandhu
- Department of Pharmacology, Weill Cornell College of Medicine, 1300 York Avenue, New York, NY, 10065, USA
| | - Faranak Fattahi
- The Center for Stem Cell Biology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, Box 256, New York, NY, 10065, USA.,Developmental Biology Program, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, Box 256, New York, NY, 10065, USA
| | - Bastian Zimmer
- The Center for Stem Cell Biology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, Box 256, New York, NY, 10065, USA.,Developmental Biology Program, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, Box 256, New York, NY, 10065, USA
| | - Smit K Shah
- Program in Chemical Biology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| | - Elizabeth Chang
- Proteomics Core Facility, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| | - Carmen Inda
- Program in Chemical Biology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA.,Hostos Community College, City University of New York, Bronx, NY, 10453, USA
| | - John Koren
- Program in Chemical Biology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA.,Department of Biochemistry, University of Notre Dame, Notre Dame, IN, 46556, USA
| | - Nathalie G Saurat
- The Center for Stem Cell Biology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, Box 256, New York, NY, 10065, USA.,Developmental Biology Program, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, Box 256, New York, NY, 10065, USA
| | - Marcel Leist
- Doerenkamp-Zbinden Chair for In Vitro Toxicology and Biomedicine, University of Konstanz, Konstanz, 78464, Germany
| | - Steven S Gross
- Department of Pharmacology, Weill Cornell College of Medicine, 1300 York Avenue, New York, NY, 10065, USA
| | - Venkatraman E Seshan
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, 10017, USA
| | - Christine Klein
- Institute of Neurogenetics, University of Lübeck, Lübeck, 23538, Germany
| | - Mark J Tomishima
- The Center for Stem Cell Biology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, Box 256, New York, NY, 10065, USA.,Developmental Biology Program, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, Box 256, New York, NY, 10065, USA.,SKI Stem Cell Research Facility, 1275 York Avenue, Sloan Kettering Institute, New York, NY, 10065, USA
| | - Hediye Erdjument-Bromage
- Department of Cell Biology, NYU School of Medicine, New York, NY, 10016, USA.,Kimmel Center for Biology and Medicine at the Skirball Institute, NYU School of Medicine, New York, NY, 10016, USA
| | - Thomas A Neubert
- Department of Cell Biology, NYU School of Medicine, New York, NY, 10016, USA.,Kimmel Center for Biology and Medicine at the Skirball Institute, NYU School of Medicine, New York, NY, 10016, USA
| | - Ronald C Henrickson
- Proteomics Core Facility, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| | - Gabriela Chiosis
- Program in Chemical Biology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA. .,Department of Medicine, Memorial Hospital, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA.
| | - Lorenz Studer
- The Center for Stem Cell Biology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, Box 256, New York, NY, 10065, USA.,Developmental Biology Program, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, Box 256, New York, NY, 10065, USA
| |
Collapse
|
56
|
de Klerk B, Emam M, Thompson-Crispi KA, Sargolzaei M, van der Poel JJ, Mallard BA. A genome-wide association study for natural antibodies measured in blood of Canadian Holstein cows. BMC Genomics 2018; 19:694. [PMID: 30241501 PMCID: PMC6150957 DOI: 10.1186/s12864-018-5062-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Accepted: 09/10/2018] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Natural antibodies (NAb) are an important component of the innate immune system, and fight infections as a part of the first line defence. NAb are poly-reactive and can respond non-specifically to antigens. Therefore, NAb may be a key trait when evaluating an animal's potential natural disease resistance. Variation in NAb is caused by both genetic and environmental factors. In this study genetic parameters of NAb were estimated and a genome-wide association study (GWAS) was performed to gain further understanding on the genes that are responsible for the observed genetic variation of NAb in Canadian Holsteins. RESULTS In total, blood samples of 1327 cows from 64 farms were studied. NAb binding to keyhole limpet hemocyanin (KLH) were determined via indirect ELISA. Immunoglobulin (Ig) isotypes, IgG and IgM, were evaluated. From the sample population, 925 cows were genotyped for 45,187 markers and each individual marker was tested to detect genetic variation in NAb levels. The relationships among animals was accounted for with genomic relationship. Results show heritabilities of 0.27 ± 0.064 (IgG) and 0.31 ± 0.065 (IgM). In total, 23 SNPs were found to be associated with IgG, but no SNPs were associated with IgM (FDR p-value < 0.05). The significant SNPs were located on autosomal chromosomes 1, 20 and 21 of the cow genome. Functional annotation analysis of the positional candidate genes revealed two sets of genes with biologically relevant functions related to NAb. In one set, seven genes with crucial roles in the production of antibody in B cells were associated with the trafficking of vesicles inside the cells between organelles. In the second set, two genes among positional candidate genes were associated with isotype class-switching and somatic hypermutation of B cells. CONCLUSIONS This study demonstrated the possibility of increasing NAb through selective breeding. In addition, the effects of two candidate pathways are proposed for further investigation of NAb production in Holsteins.
Collapse
Affiliation(s)
- Britt de Klerk
- Animal Breeding and Genomics Centre, Wageningen University, P.O. Box 338, Wageningen, The Netherlands
| | - Mehdi Emam
- Centre for Genetic Improvement of Livestock, University of Guelph, Guelph, ON, Canada.,Department of Pathobiology, Ontario Veterinary College, Genetic improvement of livestock, University of Guelph, Guelph, ON, N1G 2W1, Canada
| | | | - Mehdi Sargolzaei
- Centre for Genetic Improvement of Livestock, University of Guelph, Guelph, ON, Canada.,Semex Alliance, Guelph, ON, Canada
| | - Johan J van der Poel
- Animal Breeding and Genomics Centre, Wageningen University, P.O. Box 338, Wageningen, The Netherlands
| | - Bonnie A Mallard
- Centre for Genetic Improvement of Livestock, University of Guelph, Guelph, ON, Canada. .,Department of Pathobiology, Ontario Veterinary College, Genetic improvement of livestock, University of Guelph, Guelph, ON, N1G 2W1, Canada.
| |
Collapse
|
57
|
Gouazé-Andersson V, Ghérardi MJ, Lemarié A, Gilhodes J, Lubrano V, Arnauduc F, Cohen-Jonathan Moyal E, Toulas C. FGFR1/FOXM1 pathway: a key regulator of glioblastoma stem cells radioresistance and a prognosis biomarker. Oncotarget 2018; 9:31637-31649. [PMID: 30167084 PMCID: PMC6114977 DOI: 10.18632/oncotarget.25827] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2018] [Accepted: 07/13/2018] [Indexed: 02/07/2023] Open
Abstract
Glioblastoma are known to be aggressive and therapy-resistant tumors, due to the presence of glioblastoma stem cells inside this heterogeneous tumor. We investigate here the involvement of FGFR1 in glioblastoma stem-like cells (GSLC) radioresistance mechanisms. We first demonstrated that the survival after irradiation was significantly diminished in FGFR1-silenced (FGFR1-) GSLC compared to control GSLC. The transcriptome analysis of GSLCs FGFR1(-) showed that FOX family members are differentially regulated by FGFR1 inhibition, particularly with an upregulation of FOXN3 and a downregulation of FOXM1. GSLC survival after irradiation was significantly increased after FOXN3 silencing and decreased after FOXM1 inhibition, showing opposite effects of FGFR1/FOX family members on cell response to ionizing radiation. Silencing FGFR1 or FOXM1 downregulated genes involved in mesenchymal transition such as GLI2, TWIST1, and ZEB1 in glioblastoma stem-like cells. It also dramatically reduced GSLC migration. Databases analysis confirmed that the combined expression of FGFR1/FOXM1/MELK/GLI2/ZEB1/TWIST1 is significantly associated with patients overall survival after chemo-radiotherapy treatment. All these results, associated with our previous conduced ones with differentiated cells, clearly established that FGFR1-FOXM1 dependent glioblastoma stem-like cells radioresistance pathway is a central actor of GBM treatment resistance and a key target to inhibit in the aim to increase the sensitivity of GBM to the radiotherapy.
Collapse
Affiliation(s)
- Valérie Gouazé-Andersson
- Institut National de la Santé et de la Recherche Médicale (INSERM) UMR1037/Université Toulouse III Paul Sabatier, Cancer Research Center of Toulouse (CRCT), Toulouse, F-31000, France
| | - Marie-Julie Ghérardi
- Institut National de la Santé et de la Recherche Médicale (INSERM) UMR1037/Université Toulouse III Paul Sabatier, Cancer Research Center of Toulouse (CRCT), Toulouse, F-31000, France
| | - Anthony Lemarié
- Institut National de la Santé et de la Recherche Médicale (INSERM) UMR1037/Université Toulouse III Paul Sabatier, Cancer Research Center of Toulouse (CRCT), Toulouse, F-31000, France
| | - Julia Gilhodes
- Institut Claudius Regaud, IUCT-O, Toulouse, F-31059, France
| | - Vincent Lubrano
- CHU PURPAN-Pavillon Baudot, Place du Dr Baylac, Toulouse-Cedex 3, 31024, France
| | - Florent Arnauduc
- Institut National de la Santé et de la Recherche Médicale (INSERM) UMR1037/Université Toulouse III Paul Sabatier, Cancer Research Center of Toulouse (CRCT), Toulouse, F-31000, France
| | - Elizabeth Cohen-Jonathan Moyal
- Institut National de la Santé et de la Recherche Médicale (INSERM) UMR1037/Université Toulouse III Paul Sabatier, Cancer Research Center of Toulouse (CRCT), Toulouse, F-31000, France.,Institut Claudius Regaud, IUCT-O, Toulouse, F-31059, France
| | - Christine Toulas
- Institut National de la Santé et de la Recherche Médicale (INSERM) UMR1037/Université Toulouse III Paul Sabatier, Cancer Research Center of Toulouse (CRCT), Toulouse, F-31000, France.,Institut Claudius Regaud, IUCT-O, Toulouse, F-31059, France
| |
Collapse
|
58
|
Lin Y, van Duyvenvoorde HA, Liu H, Yang C, Warsito D, Yin C, Kant SG, Haglund F, Wit JM, Larsson O. Characterization of an activating R1353H insulin-like growth factor 1 receptor variant in a male with extreme tall height. Eur J Endocrinol 2018; 179:85-95. [PMID: 29789409 DOI: 10.1530/eje-18-0176] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Accepted: 05/22/2018] [Indexed: 11/08/2022]
Abstract
OBJECTIVE The insulin-like growth factor1 receptor (IGF1R) is important in growth and development, and inactivating IGF1R mutations cause short stature and relatively high levels of serum IGF-I. We identified an unclassified IGF1RR1353H variant in a male with extreme tall height, very low levels of serum IGF-I and delayed and prolonged growth spurt. The index case's mother and three sons all carried the variant, but so far only the eldest son (age 18 years) presented with tall height. We hypothesized that the variant could constitute an activating mutation. DESIGN The IGF1RR1353H variant was investigated in Igf1r-/- mouse embryonic fibroblasts (R-cells) by cell cycle, colony formation and transcriptome analyses. RESULTS The IGF1RR1353H (R-1353) exhibited significantly increased cell proliferation, G1-S progression and colony formation in soft agar. RNA sequencing identified 195 differentially expressed genes between R-WT and R-1353 (adjusted P < 1E-100). Most genes were upregulated in R-1353, including the gene encoding the androgen receptor (AR). Gene expression profiling showed the most significant enrichment in extracellular matrix organization (P = 2.76E-7), collagen biosynthesis (P = 1.21E-5) and cell adhesion (P = 7.38E-5). Retrospective biochemical analysis of the index case revealed decreased testosterone and sex hormone-binding globulin levels, whereas LH and FSH were within normal ranges. This profile suggests an increased sensitivity to androgen, which is compatible with the enhanced expression of Ar in R-1353 cells. CONCLUSIONS Our findings suggest that R1353H constitutes an activating IGF1R variant. The possible deregulation of collagen turnover and increased androgen sensitivity implicates an association to tall phenotype in male carriers.
Collapse
MESH Headings
- Adult
- Amino Acid Substitution
- Animals
- Body Height
- Cell Line
- Cell Proliferation
- Down-Regulation
- Gene Expression Regulation, Developmental
- Green Fluorescent Proteins/chemistry
- Green Fluorescent Proteins/genetics
- Green Fluorescent Proteins/metabolism
- Growth Disorders/blood
- Growth Disorders/genetics
- Growth Disorders/metabolism
- Growth Disorders/physiopathology
- Heterozygote
- Humans
- Insulin-Like Growth Factor I/analysis
- Male
- Mice, Knockout
- Pedigree
- Point Mutation
- RNA, Messenger/chemistry
- RNA, Messenger/metabolism
- Receptor, IGF Type 1/chemistry
- Receptor, IGF Type 1/genetics
- Receptor, IGF Type 1/metabolism
- Receptors, Androgen/genetics
- Receptors, Androgen/metabolism
- Recombinant Fusion Proteins/metabolism
- Sequence Analysis, RNA
- Severity of Illness Index
Collapse
Affiliation(s)
- Yingbo Lin
- Department of Oncology-Pathology, Karolinska Institutet, Cancer Center Karolinska (CCK), Karolinska University Hospital, Stockholm, Sweden
| | | | - Hongyu Liu
- Department of Oncology-Pathology, Karolinska Institutet, Cancer Center Karolinska (CCK), Karolinska University Hospital, Stockholm, Sweden
- Laboratory of Aquatic Animal Nutrition and Feed, Fisheries College, Guangdong Ocean University, Zhanjiang, China
| | - Chen Yang
- Department of Oncology-Pathology, Karolinska Institutet, Cancer Center Karolinska (CCK), Karolinska University Hospital, Stockholm, Sweden
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Dudi Warsito
- Department of Oncology-Pathology, Karolinska Institutet, Cancer Center Karolinska (CCK), Karolinska University Hospital, Stockholm, Sweden
| | - Chang Yin
- Department of Oncology-Pathology, Karolinska Institutet, Cancer Center Karolinska (CCK), Karolinska University Hospital, Stockholm, Sweden
| | - Sarina G Kant
- Department of Clinical Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Felix Haglund
- Department of Oncology-Pathology, Karolinska Institutet, Cancer Center Karolinska (CCK), Karolinska University Hospital, Stockholm, Sweden
| | - Jan M Wit
- Department of Paediatrics, Leiden University Medical Center, Leiden, The Netherlands
| | - Olle Larsson
- Department of Oncology-Pathology, Karolinska Institutet, Cancer Center Karolinska (CCK), Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
59
|
Semantic Analysis of Posttranslational Modification of Proteins Accumulated in Thyroid Cancer Cells Exposed to Simulated Microgravity. Int J Mol Sci 2018; 19:ijms19082257. [PMID: 30071661 PMCID: PMC6121900 DOI: 10.3390/ijms19082257] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 07/27/2018] [Accepted: 07/30/2018] [Indexed: 02/08/2023] Open
Abstract
When monolayers of tissue cancer cells of various origins are exposed to real or simulated microgravity, many cells leave the monolayer and assemble to three-dimensional (3D) aggregates (spheroids). In order to define the cellular machinery leading to this change in growth behavior of FTC-133 human thyroid cancer cells and MCF-7 breast cancer cells, we recently performed proteome analyses on these cell lines and determined the proteins’ accumulation in monolayer cells grown under 1g-conditions as well as in the cells of spheroids assembled under simulated microgravity during three and 14 days, respectively. At that time, an influence of the increment or decrement of some of the more than 5000 proteins detected in each cell line was investigated. In this study, we focused on posttranslational modifications (PTMs) of proteins. For this purpose, we selected candidates from the list of the proteins detected in the two preceding proteome analyses, which showed significant accumulation in spheroid cells as compared to 1g monolayer cells. Then we searched for those PTMs of the selected proteins, which according to the literature have already been determined experimentally. Using the Semantic Protocol and RDF Query Language (SPARQL), various databases were examined. Most efficient was the search in the latest version of the dbPTM database. In total, we found 72 different classes of PTMs comprising mainly phosphorylation, glycosylation, ubiquitination and acetylation. Most interestingly, in 35 of the 69 proteins, N6 residues of lysine are modifiable.
Collapse
|
60
|
Shan X, Roberts C, Lan Y, Percec I. Age Alters Chromatin Structure and Expression of SUMO Proteins under Stress Conditions in Human Adipose-Derived Stem Cells. Sci Rep 2018; 8:11502. [PMID: 30065345 PMCID: PMC6068198 DOI: 10.1038/s41598-018-29775-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Accepted: 07/14/2018] [Indexed: 12/19/2022] Open
Abstract
Adult stem cells play a critical role in tissue homeostasis and repair. Aging leads to a decline in stem cells’ regenerative capacity that contributes significantly to the maintenance of organ and tissue functions. Age-dependent genomic and epigenetic modifications together play a role in the disruption of critical cellular pathways. However, the epigenetic mechanisms responsible for the decline of adult stem cell functions remain to be well established. Here, we investigated age-dependent, genome-wide alterations in the chromatin accessibility of primary human adipose-derived stem cells (ASCs) in comparison to age-matched fibroblasts via ATAC-seq technology. Our results demonstrate that aging ASCs possess globally more stable chromatin accessibility profiles as compared to aging fibroblasts, suggesting that robust regulatory mechanisms maintain adult stem cell chromatin structure against aging. Furthermore, we observed age-dependent subtle changes in promoter nucleosome positioning in selective pathways during aging, concurrent with altered small ubiquitin-related modifier (SUMO) protein expression under stress conditions. Together, our data suggest a significant role for nucleosome positioning in sumoylation pathway regulation in stress response during adult stem cell aging. The differences described here between the chromatin structure of human ASCs and fibroblasts will further elucidate the mechanisms regulating gene expression during aging in both stem cells and differentiated cells.
Collapse
Affiliation(s)
- Xiaoyin Shan
- Department of Surgery, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Cleresa Roberts
- Department of Surgery, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Yemin Lan
- Epigenetics Institute, Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Ivona Percec
- Department of Surgery, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| |
Collapse
|
61
|
Flitcroft DI, Loughman J, Wildsoet CF, Williams C, Guggenheim JA. Novel Myopia Genes and Pathways Identified From Syndromic Forms of Myopia. Invest Ophthalmol Vis Sci 2018; 59:338-348. [PMID: 29346494 PMCID: PMC5773233 DOI: 10.1167/iovs.17-22173] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Purpose To test the hypothesis that genes known to cause clinical syndromes featuring myopia also harbor polymorphisms contributing to nonsyndromic refractive errors. Methods Clinical phenotypes and syndromes that have refractive errors as a recognized feature were identified using the Online Mendelian Inheritance in Man (OMIM) database. One hundred fifty-four unique causative genes were identified, of which 119 were specifically linked with myopia and 114 represented syndromic myopia (i.e., myopia and at least one other clinical feature). Myopia was the only refractive error listed for 98 genes and hyperopia and the only refractive error noted for 28 genes, with the remaining 28 genes linked to phenotypes with multiple forms of refractive error. Pathway analysis was carried out to find biological processes overrepresented within these sets of genes. Genetic variants located within 50 kb of the 119 myopia-related genes were evaluated for involvement in refractive error by analysis of summary statistics from genome-wide association studies (GWAS) conducted by the CREAM Consortium and 23andMe, using both single-marker and gene-based tests. Results Pathway analysis identified several biological processes already implicated in refractive error development through prior GWAS analyses and animal studies, including extracellular matrix remodeling, focal adhesion, and axon guidance, supporting the research hypothesis. Novel pathways also implicated in myopia development included mannosylation, glycosylation, lens development, gliogenesis, and Schwann cell differentiation. Hyperopia was found to be linked to a different pattern of biological processes, mostly related to organogenesis. Comparison with GWAS findings further confirmed that syndromic myopia genes were enriched for genetic variants that influence refractive errors in the general population. Gene-based analyses implicated 21 novel candidate myopia genes (ADAMTS18, ADAMTS2, ADAMTSL4, AGK, ALDH18A1, ASXL1, COL4A1, COL9A2, ERBB3, FBN1, GJA1, GNPTG, IFIH1, KIF11, LTBP2, OCA2, POLR3B, POMT1, PTPN11, TFAP2A, ZNF469). Conclusions Common genetic variants within or nearby genes that cause syndromic myopia are enriched for variants that cause nonsyndromic, common myopia. Analysis of syndromic forms of refractive errors can provide new insights into the etiology of myopia and additional potential targets for therapeutic interventions.
Collapse
Affiliation(s)
- D Ian Flitcroft
- Children's University Hospital and University College Dublin, Dublin, Ireland.,College of Sciences and Health, Dublin Institute of Technology, Dublin, Ireland
| | - James Loughman
- College of Sciences and Health, Dublin Institute of Technology, Dublin, Ireland
| | - Christine F Wildsoet
- Center for Eye Disease and Development, School of Optometry, University of California-Berkeley, Berkeley, California, United States
| | - Cathy Williams
- Bristol Eye Hospital and Bristol University, Bristol, United Kingdom
| | - Jeremy A Guggenheim
- School of Optometry & Vision Sciences, Cardiff University, Cardiff, United Kingdom
| | | |
Collapse
|
62
|
Jung H, Choi JK, Lee EA. Immune signatures correlate with L1 retrotransposition in gastrointestinal cancers. Genome Res 2018; 28:1136-1146. [PMID: 29970450 PMCID: PMC6071633 DOI: 10.1101/gr.231837.117] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Accepted: 06/29/2018] [Indexed: 12/15/2022]
Abstract
Long interspersed nuclear element-1 (LINE-1 or L1) retrotransposons are normally suppressed in somatic tissues mainly due to DNA methylation and antiviral defense. However, the mechanism to suppress L1s may be disrupted in cancers, thus allowing L1s to act as insertional mutagens and cause genomic rearrangement and instability. Whereas the frequency of somatic L1 insertions varies greatly among individual tumors, much remains to be learned about underlying genetic, cellular, or environmental factors. Here, we report multiple correlates of L1 activity in stomach, colorectal, and esophageal tumors through an integrative analysis of cancer whole-genome and matched RNA-sequencing profiles. Clinical indicators of tumor progression, such as tumor grade and patient age, showed positive association. A potential L1 expression suppressor, TP53, was mutated in tumors with frequent L1 insertions. We characterized the effects of somatic L1 insertions on mRNA splicing and expression, and demonstrated an increased risk of gene disruption in retrotransposition-prone cancers. In particular, we found that a cancer-specific L1 insertion in an exon of MOV10, a key L1 suppressor, caused exon skipping and decreased expression of the affected allele due to nonsense-mediated decay in a tumor with a high L1 insertion load. Importantly, tumors with high immune activity, for example, those associated with Epstein–Barr virus infection or microsatellite instability, tended to carry a low number of L1 insertions in genomes with high expression levels of L1 suppressors such as APOBEC3s and SAMHD1. Our results indicate that cancer immunity may contribute to genome stability by suppressing L1 retrotransposition in gastrointestinal cancers.
Collapse
Affiliation(s)
- Hyunchul Jung
- Department of Bio and Brain Engineering, KAIST, Daejeon 34141, South Korea
| | - Jung Kyoon Choi
- Department of Bio and Brain Engineering, KAIST, Daejeon 34141, South Korea
| | - Eunjung Alice Lee
- Division of Genetics and Genomics, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts 02115, USA
| |
Collapse
|
63
|
Li C, Rezov V, Joensuu E, Vartiainen V, Rönty M, Yin M, Myllärniemi M, Koli K. Pirfenidone decreases mesothelioma cell proliferation and migration via inhibition of ERK and AKT and regulates mesothelioma tumor microenvironment in vivo. Sci Rep 2018; 8:10070. [PMID: 29968778 PMCID: PMC6030186 DOI: 10.1038/s41598-018-28297-x] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Accepted: 06/13/2018] [Indexed: 12/29/2022] Open
Abstract
Malignant mesothelioma is an aggressive cancer with poor prognosis. It is characterized by prominent extracellular matrix, mesenchymal tumor cell phenotypes and chemoresistance. In this study, the ability of pirfenidone to alter mesothelioma cell proliferation and migration as well as mesothelioma tumor microenvironment was evaluated. Pirfenidone is an anti-fibrotic drug used in the treatment of idiopathic pulmonary fibrosis and has also anti-proliferative activities. Mesothelioma cell proliferation was decreased by pirfenidone alone or in combination with cisplatin. Pirfenidone also decreased significantly Transwell migration/invasion and 3D collagen invasion. This was associated with increased BMP pathway activity, decreased GREM1 expression and downregulation of MAPK/ERK and AKT/mTOR signaling. The canonical Smad-mediated TGF-β signaling was not affected by pirfenidone. However, pirfenidone blocked TGF-β induced upregulation of ERK and AKT pathways. Treatment of mice harboring mesothelioma xenografts with pirfenidone alone did not reduce tumor proliferation in vivo. However, pirfenidone modified the tumor microenvironment by reducing the expression of extracellular matrix associated genes. In addition, GREM1 expression was downregulated by pirfenidone in vivo. By reducing two major upregulated pathways in mesothelioma and by targeting tumor cells and the microenvironment pirfenidone may present a novel anti-fibrotic and anti-cancer adjuvant therapy for mesothelioma.
Collapse
Affiliation(s)
- Chang Li
- Research Programs Unit, Translational Cancer Biology, University of Helsinki, Helsinki, Finland.,Department of Thoracic and Cardiovascular Surgery, The First Affiliated Hospital of Soochow University, Medical College of Soochow University, Soochow, China
| | - Veronika Rezov
- Research Programs Unit, Translational Cancer Biology, University of Helsinki, Helsinki, Finland
| | - Emmi Joensuu
- Research Programs Unit, Translational Cancer Biology, University of Helsinki, Helsinki, Finland
| | - Ville Vartiainen
- Research Programs Unit, Translational Cancer Biology, University of Helsinki, Helsinki, Finland.,University of Helsinki and Helsinki University Hospital, Heart and Lung Center and HUH diagnostics, Pulmonary Medicine, Helsinki, Finland
| | - Mikko Rönty
- Department of Pathology, University of Helsinki and Fimlab laboratories, Pathology, Tampere, Finland
| | - Miao Yin
- Research Programs Unit, Translational Cancer Biology, University of Helsinki, Helsinki, Finland
| | - Marjukka Myllärniemi
- University of Helsinki and Helsinki University Hospital, Heart and Lung Center and HUH diagnostics, Pulmonary Medicine, Helsinki, Finland
| | - Katri Koli
- Research Programs Unit, Translational Cancer Biology, University of Helsinki, Helsinki, Finland.
| |
Collapse
|
64
|
Sahana J, Nassef MZ, Wehland M, Kopp S, Krüger M, Corydon TJ, Infanger M, Bauer J, Grimm D. Decreased E-Cadherin in MCF7 Human Breast Cancer Cells Forming Multicellular Spheroids Exposed to Simulated Microgravity. Proteomics 2018; 18:e1800015. [PMID: 29785723 DOI: 10.1002/pmic.201800015] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Revised: 04/27/2018] [Indexed: 12/23/2022]
Abstract
MCF7 human breast cancer cells were cultured under normal gravity (1 g) and on a random positioning machine (RPM) preventing sedimentation. After 2 weeks, adherent 1 g-control and adherent RPM cells (AD) as well as multicellular spheroids (MCS) were harvested. AD and MCS had been exposed to the RPM in the same culture flask. In a subsequent proteome analysis, the majority of the proteins detected showed similar label-free quantification (LFQ) scores in each of the respective subpopulations, but in both AD or MCS cultures, proteins were also found whose LFQs deviated at least twofold from their counterparts in the 1 g-control cells. They included the cell junction protein E-cadherin, which was diminished in MCS cells, where proteins of the E-cadherin autodegradation pathway were enhanced and c-Src (proto-oncogene tyrosine-protein kinase c-Src) was detected. Spheroid formation was prevented by inhibition of c-Src but promoted by antibodies blocking E-cadherin activity. An interaction analysis of the detected proteins that are involved in forming and regulating junctions or adhesion complexes and in E-cadherin autodegradation indicated connections between the two protein groups. This suggests that the balance of proteins that up- or downregulate E-cadherin mediates the tendency of MCF7 cells to form MCS during RPM exposure.
Collapse
Affiliation(s)
- Jayashree Sahana
- Department of Biomedicine, Aarhus University, 8000 Aarhus C, Denmark
| | - Mohamed Zakaria Nassef
- Clinic for Plastic, Aesthetic and Hand Surgery, Otto-von-Guericke-University Magdeburg, 39120 Magdeburg, Germany
| | - Markus Wehland
- Clinic for Plastic, Aesthetic and Hand Surgery, Otto-von-Guericke-University Magdeburg, 39120 Magdeburg, Germany
| | - Sascha Kopp
- Clinic for Plastic, Aesthetic and Hand Surgery, Otto-von-Guericke-University Magdeburg, 39120 Magdeburg, Germany
| | - Marcus Krüger
- Clinic for Plastic, Aesthetic and Hand Surgery, Otto-von-Guericke-University Magdeburg, 39120 Magdeburg, Germany
| | - Thomas J Corydon
- Department of Biomedicine, Aarhus University, 8000 Aarhus C, Denmark.,Department of Ophthalmology, Aarhus University Hospital, 8000 Aarhus C, Denmark
| | - Manfred Infanger
- Clinic for Plastic, Aesthetic and Hand Surgery, Otto-von-Guericke-University Magdeburg, 39120 Magdeburg, Germany
| | - Johann Bauer
- Max-Planck Institute of Biochemistry, 82152 Martinsried, Germany
| | - Daniela Grimm
- Department of Biomedicine, Aarhus University, 8000 Aarhus C, Denmark.,Clinic for Plastic, Aesthetic and Hand Surgery, Otto-von-Guericke-University Magdeburg, 39120 Magdeburg, Germany.,Gravitational Biology and Translational Regenerative Medicine, Faculty of Medicine and Mechanical Engineering, Otto-von-Guericke-University Magdeburg, 39120 Magdeburg, Germany
| |
Collapse
|
65
|
Linking transcriptional and genetic tumor heterogeneity through allele analysis of single-cell RNA-seq data. Genome Res 2018; 28:1217-1227. [PMID: 29898899 PMCID: PMC6071640 DOI: 10.1101/gr.228080.117] [Citation(s) in RCA: 117] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2017] [Accepted: 05/24/2018] [Indexed: 12/27/2022]
Abstract
Characterization of intratumoral heterogeneity is critical to cancer therapy, as the presence of phenotypically diverse cell populations commonly fuels relapse and resistance to treatment. Although genetic variation is a well-studied source of intratumoral heterogeneity, the functional impact of most genetic alterations remains unclear. Even less understood is the relative importance of other factors influencing heterogeneity, such as epigenetic state or tumor microenvironment. To investigate the relationship between genetic and transcriptional heterogeneity in a context of cancer progression, we devised a computational approach called HoneyBADGER to identify copy number variation and loss of heterozygosity in individual cells from single-cell RNA-sequencing data. By integrating allele and normalized expression information, HoneyBADGER is able to identify and infer the presence of subclone-specific alterations in individual cells and reconstruct the underlying subclonal architecture. By examining several tumor types, we show that HoneyBADGER is effective at identifying deletions, amplifications, and copy-neutral loss-of-heterozygosity events and is capable of robustly identifying subclonal focal alterations as small as 10 megabases. We further apply HoneyBADGER to analyze single cells from a progressive multiple myeloma patient to identify major genetic subclones that exhibit distinct transcriptional signatures relevant to cancer progression. Other prominent transcriptional subpopulations within these tumors did not line up with the genetic subclonal structure and were likely driven by alternative, nonclonal mechanisms. These results highlight the need for integrative analysis to understand the molecular and phenotypic heterogeneity in cancer.
Collapse
|
66
|
Bazzazi H, Zhang Y, Jafarnejad M, Isenberg JS, Annex BH, Popel AS. Computer Simulation of TSP1 Inhibition of VEGF-Akt-eNOS: An Angiogenesis Triple Threat. Front Physiol 2018; 9:644. [PMID: 29899706 PMCID: PMC5988849 DOI: 10.3389/fphys.2018.00644] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Accepted: 05/11/2018] [Indexed: 01/08/2023] Open
Abstract
The matricellular protein thrombospondin-1 (TSP1) is a potent inhibitor of angiogenesis. Specifically, TSP1 has been experimentally shown to inhibit signaling downstream of vascular endothelial growth factor (VEGF). The molecular mechanism of this inhibition is not entirely clear. We developed a detailed computational model of VEGF signaling to Akt-endothelial nitric oxide synthase (eNOS) to investigate the quantitative molecular mechanism of TSP1 inhibition. The model demonstrated that TSP1 acceleration of VEGFR2 degradation is sufficient to explain the inhibition of VEGFR2 and eNOS phosphorylation. However, Akt inhibition requires TSP1-induced phosphatase recruitment to VEGFR2. The model was then utilized to test various strategies for the rescue of VEGF signaling to Akt and eNOS. Inhibiting TSP1 was predicted to be not as effective as CD47 depletion in rescuing signaling to Akt. The model further predicts that combination strategy involving depletion of CD47 and inhibition of TSP1 binding to CD47 is necessary for effective recovery of signaling to eNOS. In all, computational modeling offers insight to molecular mechanisms involving TSP1 interaction with VEGF signaling and provides strategies for rescuing angiogenesis by targeting TSP1-CD47 axis.
Collapse
Affiliation(s)
- Hojjat Bazzazi
- Department of Biomedical Engineering, School of Medicine, Johns Hopkins University, Baltimore, MD, United States
| | - Yu Zhang
- Department of Biomedical Engineering, School of Medicine, Johns Hopkins University, Baltimore, MD, United States
| | - Mohammad Jafarnejad
- Department of Biomedical Engineering, School of Medicine, Johns Hopkins University, Baltimore, MD, United States
| | - Jeffrey S Isenberg
- Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, United States.,Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Brian H Annex
- Division of Cardiovascular Medicine, Department of Medicine, Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA, United States
| | - Aleksander S Popel
- Department of Biomedical Engineering, School of Medicine, Johns Hopkins University, Baltimore, MD, United States
| |
Collapse
|
67
|
De Meulder B, Lefaudeux D, Bansal AT, Mazein A, Chaiboonchoe A, Ahmed H, Balaur I, Saqi M, Pellet J, Ballereau S, Lemonnier N, Sun K, Pandis I, Yang X, Batuwitage M, Kretsos K, van Eyll J, Bedding A, Davison T, Dodson P, Larminie C, Postle A, Corfield J, Djukanovic R, Chung KF, Adcock IM, Guo YK, Sterk PJ, Manta A, Rowe A, Baribaud F, Auffray C. A computational framework for complex disease stratification from multiple large-scale datasets. BMC SYSTEMS BIOLOGY 2018; 12:60. [PMID: 29843806 PMCID: PMC5975674 DOI: 10.1186/s12918-018-0556-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Accepted: 02/21/2018] [Indexed: 01/05/2023]
Abstract
BACKGROUND Multilevel data integration is becoming a major area of research in systems biology. Within this area, multi-'omics datasets on complex diseases are becoming more readily available and there is a need to set standards and good practices for integrated analysis of biological, clinical and environmental data. We present a framework to plan and generate single and multi-'omics signatures of disease states. METHODS The framework is divided into four major steps: dataset subsetting, feature filtering, 'omics-based clustering and biomarker identification. RESULTS We illustrate the usefulness of this framework by identifying potential patient clusters based on integrated multi-'omics signatures in a publicly available ovarian cystadenocarcinoma dataset. The analysis generated a higher number of stable and clinically relevant clusters than previously reported, and enabled the generation of predictive models of patient outcomes. CONCLUSIONS This framework will help health researchers plan and perform multi-'omics big data analyses to generate hypotheses and make sense of their rich, diverse and ever growing datasets, to enable implementation of translational P4 medicine.
Collapse
Affiliation(s)
- Bertrand De Meulder
- European Institute for Systems Biology and Medicine, CNRS-ENS-UCBL, EISBM, 50 Avenue Tony Garnier, 69007, Lyon, France.
| | - Diane Lefaudeux
- European Institute for Systems Biology and Medicine, CNRS-ENS-UCBL, EISBM, 50 Avenue Tony Garnier, 69007, Lyon, France
| | - Aruna T Bansal
- Acclarogen Ltd, St John's Innovation Centre, Cambridge, CB4 OWS, UK
| | - Alexander Mazein
- European Institute for Systems Biology and Medicine, CNRS-ENS-UCBL, EISBM, 50 Avenue Tony Garnier, 69007, Lyon, France
| | - Amphun Chaiboonchoe
- European Institute for Systems Biology and Medicine, CNRS-ENS-UCBL, EISBM, 50 Avenue Tony Garnier, 69007, Lyon, France
| | - Hassan Ahmed
- European Institute for Systems Biology and Medicine, CNRS-ENS-UCBL, EISBM, 50 Avenue Tony Garnier, 69007, Lyon, France
| | - Irina Balaur
- European Institute for Systems Biology and Medicine, CNRS-ENS-UCBL, EISBM, 50 Avenue Tony Garnier, 69007, Lyon, France
| | - Mansoor Saqi
- European Institute for Systems Biology and Medicine, CNRS-ENS-UCBL, EISBM, 50 Avenue Tony Garnier, 69007, Lyon, France
| | - Johann Pellet
- European Institute for Systems Biology and Medicine, CNRS-ENS-UCBL, EISBM, 50 Avenue Tony Garnier, 69007, Lyon, France
| | - Stéphane Ballereau
- European Institute for Systems Biology and Medicine, CNRS-ENS-UCBL, EISBM, 50 Avenue Tony Garnier, 69007, Lyon, France
| | - Nathanaël Lemonnier
- European Institute for Systems Biology and Medicine, CNRS-ENS-UCBL, EISBM, 50 Avenue Tony Garnier, 69007, Lyon, France
| | - Kai Sun
- Data Science Institute, Imperial College, London, SW7 2AZ, UK
| | - Ioannis Pandis
- Data Science Institute, Imperial College, London, SW7 2AZ, UK.,Janssen Research and Development Ltd, High Wycombe, HP12 4DP, UK
| | - Xian Yang
- Data Science Institute, Imperial College, London, SW7 2AZ, UK
| | | | | | | | | | - Timothy Davison
- Janssen Research and Development Ltd, High Wycombe, HP12 4DP, UK
| | - Paul Dodson
- AstraZeneca Ltd, Alderley Park, Macclesfield, SK10 4TG, UK
| | | | - Anthony Postle
- Faculty of Medicine, University of Southampton, Southampton, SO17 1BJ, UK
| | - Julie Corfield
- AstraZeneca R & D, 43150, Mölndal, Sweden.,Arateva R & D Ltd, Nottingham, NG1 1GF, UK
| | - Ratko Djukanovic
- Faculty of Medicine, University of Southampton, Southampton, SO17 1BJ, UK
| | - Kian Fan Chung
- National Hearth and Lung Institute, Imperial College London, London, SW3 6LY, UK
| | - Ian M Adcock
- National Hearth and Lung Institute, Imperial College London, London, SW3 6LY, UK
| | - Yi-Ke Guo
- Data Science Institute, Imperial College, London, SW7 2AZ, UK
| | - Peter J Sterk
- Department of Respiratory Medicine, Academic Medical Centre, University of Amsterdam, Amsterdam, AZ1105, The Netherlands
| | - Alexander Manta
- Research Informatics, Roche Diagnostics GmbH, 82008, Unterhaching, Germany
| | - Anthony Rowe
- Janssen Research and Development Ltd, High Wycombe, HP12 4DP, UK
| | | | - Charles Auffray
- European Institute for Systems Biology and Medicine, CNRS-ENS-UCBL, EISBM, 50 Avenue Tony Garnier, 69007, Lyon, France.
| | | |
Collapse
|
68
|
Mistimed food intake and sleep alters 24-hour time-of-day patterns of the human plasma proteome. Proc Natl Acad Sci U S A 2018; 115:E5390-E5399. [PMID: 29784788 DOI: 10.1073/pnas.1714813115] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Proteomics holds great promise for understanding human physiology, developing health biomarkers, and precision medicine. However, how much the plasma proteome varies with time of day and is regulated by the master circadian suprachiasmatic nucleus brain clock, assessed here by the melatonin rhythm, is largely unknown. Here, we assessed 24-h time-of-day patterns of human plasma proteins in six healthy men during daytime food intake and nighttime sleep in phase with the endogenous circadian clock (i.e., circadian alignment) versus daytime sleep and nighttime food intake out of phase with the endogenous circadian clock (i.e., circadian misalignment induced by simulated nightshift work). We identified 24-h time-of-day patterns in 573 of 1,129 proteins analyzed, with 30 proteins showing strong regulation by the circadian cycle. Relative to circadian alignment, the average abundance and/or 24-h time-of-day patterns of 127 proteins were altered during circadian misalignment. Altered proteins were associated with biological pathways involved in immune function, metabolism, and cancer. Of the 30 circadian-regulated proteins, the majority peaked between 1400 hours and 2100 hours, and these 30 proteins were associated with basic pathways involved in extracellular matrix organization, tyrosine kinase signaling, and signaling by receptor tyrosine-protein kinase erbB-2. Furthermore, circadian misalignment altered multiple proteins known to regulate glucose homeostasis and/or energy metabolism, with implications for altered metabolic physiology. Our findings demonstrate the circadian clock, the behavioral wake-sleep/food intake-fasting cycle, and interactions between these processes regulate 24-h time-of-day patterns of human plasma proteins and help identify mechanisms of circadian misalignment that may contribute to metabolic dysregulation.
Collapse
|
69
|
Liem DA, Murali S, Sigdel D, Shi Y, Wang X, Shen J, Choi H, Caufield JH, Wang W, Ping P, Han J. Phrase mining of textual data to analyze extracellular matrix protein patterns across cardiovascular disease. Am J Physiol Heart Circ Physiol 2018; 315:H910-H924. [PMID: 29775406 DOI: 10.1152/ajpheart.00175.2018] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Extracellular matrix (ECM) proteins have been shown to play important roles regulating multiple biological processes in an array of organ systems, including the cardiovascular system. Using a novel bioinformatics text-mining tool, we studied six categories of cardiovascular disease (CVD), namely, ischemic heart disease, cardiomyopathies, cerebrovascular accident, congenital heart disease, arrhythmias, and valve disease, anticipating novel ECM protein-disease and protein-protein relationships hidden within vast quantities of textual data. We conducted a phrase-mining analysis, delineating the relationships of 709 ECM proteins with the 6 groups of CVDs reported in 1,099,254 abstracts. The technology pipeline known as Context-Aware Semantic Online Analytical Processing was applied to semantically rank the association of proteins to each CVD and all six CVDs, performing analyses to quantify each protein-disease relationship. We performed principal component analysis and hierarchical clustering of the data, where each protein was visualized as a six-dimensional vector. We found that ECM proteins display variable degrees of association with the six CVDs; certain CVDs share groups of associated proteins, whereas others have divergent protein associations. We identified 82 ECM proteins sharing associations with all 6 CVDs. Our bioinformatics analysis ascribed distinct ECM pathways (via Reactome) from this subset of proteins, namely, insulin-like growth factor regulation and interleukin-4 and interleukin-13 signaling, suggesting their contribution to the pathogenesis of all six CVDs. Finally, we performed hierarchical clustering analysis and identified protein clusters predominantly associated with a targeted CVD; analyses of these proteins revealed unexpected insights underlying the key ECM-related molecular pathogenesis of each CVD, including virus assembly and release in arrhythmias. NEW & NOTEWORTHY The present study is the first application of a text-mining algorithm to characterize the relationships of 709 extracellular matrix-related proteins with 6 categories of cardiovascular disease described in 1,099,254 abstracts. Our analysis informed unexpected extracellular matrix functions, pathways, and molecular relationships implicated in the six cardiovascular diseases.
Collapse
Affiliation(s)
- David A Liem
- NIH BD2K Program Centers of Excellence for Big Data Computing-Heart BD2K Center, Departments of Physiology, Medicine/Cardiology, and Bioinformatics, David Geffen School of Medicine, University of California , Los Angeles, California
| | - Sanjana Murali
- NIH BD2K Program Centers of Excellence for Big Data Computing-Heart BD2K Center, Departments of Physiology, Medicine/Cardiology, and Bioinformatics, David Geffen School of Medicine, University of California , Los Angeles, California
| | - Dibakar Sigdel
- NIH BD2K Program Centers of Excellence for Big Data Computing-Heart BD2K Center, Departments of Physiology, Medicine/Cardiology, and Bioinformatics, David Geffen School of Medicine, University of California , Los Angeles, California
| | - Yu Shi
- NIH BD2K Program Centers of Excellence for Big Data Computing-KnowEng Center, Department of Computer Science, University of Illinois at Urbana-Champaign , Champaign, Illinois
| | - Xuan Wang
- NIH BD2K Program Centers of Excellence for Big Data Computing-KnowEng Center, Department of Computer Science, University of Illinois at Urbana-Champaign , Champaign, Illinois
| | - Jiaming Shen
- NIH BD2K Program Centers of Excellence for Big Data Computing-KnowEng Center, Department of Computer Science, University of Illinois at Urbana-Champaign , Champaign, Illinois
| | - Howard Choi
- NIH BD2K Program Centers of Excellence for Big Data Computing-Heart BD2K Center, Departments of Physiology, Medicine/Cardiology, and Bioinformatics, David Geffen School of Medicine, University of California , Los Angeles, California
| | - John H Caufield
- NIH BD2K Program Centers of Excellence for Big Data Computing-Heart BD2K Center, Departments of Physiology, Medicine/Cardiology, and Bioinformatics, David Geffen School of Medicine, University of California , Los Angeles, California
| | - Wei Wang
- NIH BD2K Program Centers of Excellence for Big Data Computing-Heart BD2K Center, Heart Big Data to Knowledge Center, Department of Computer Science, Scalable Analytics Institute, Henry Samueli School of Engineering and Applied Science, University of California , Los Angeles, California
| | - Peipei Ping
- NIH BD2K Program Centers of Excellence for Big Data Computing-Heart BD2K Center, Departments of Physiology, Medicine/Cardiology, and Bioinformatics, David Geffen School of Medicine, University of California , Los Angeles, California.,NIH BD2K Program Centers of Excellence for Big Data Computing-Heart BD2K Center, Heart Big Data to Knowledge Center, Department of Computer Science, Scalable Analytics Institute, Henry Samueli School of Engineering and Applied Science, University of California , Los Angeles, California
| | - JiaWei Han
- NIH BD2K Program Centers of Excellence for Big Data Computing-KnowEng Center, Department of Computer Science, University of Illinois at Urbana-Champaign , Champaign, Illinois
| |
Collapse
|
70
|
Mestres AC, Bochkina N, Mayer C. Selection of the Regularization Parameter in Graphical Models Using Network Characteristics. J Comput Graph Stat 2018. [DOI: 10.1080/10618600.2017.1366910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Affiliation(s)
- Adria Caballe Mestres
- University of Edinburgh & Maxwell Institute, Scotland, United Kingdom
- Biomathematics & Statistics Scotland, Scotland, United Kingdom
| | - Natalia Bochkina
- University of Edinburgh & Maxwell Institute, Scotland, United Kingdom
- The Alan Turing Institute for Data Science, British Library, London, United Kingdom
| | - Claus Mayer
- Biomathematics & Statistics Scotland, Scotland, United Kingdom
| |
Collapse
|
71
|
Pavan S, Meyer-Schaller N, Diepenbruck M, Kalathur RKR, Saxena M, Christofori G. A kinome-wide high-content siRNA screen identifies MEK5-ERK5 signaling as critical for breast cancer cell EMT and metastasis. Oncogene 2018; 37:4197-4213. [PMID: 29713055 DOI: 10.1038/s41388-018-0270-8] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Revised: 01/10/2018] [Accepted: 01/16/2018] [Indexed: 12/21/2022]
Abstract
An epithelial to mesenchymal transition (EMT) has been correlated to malignant tumor progression and metastasis by promoting cancer cell migration and invasion and chemoresistance. Hence, finding druggable EMT effectors is critical to efficiently interfere with metastasis formation and to overcome therapy resistance. We have employed a high-content microscopy screen in combination with a kinome and phosphatome-wide siRNA library to identify signaling pathways underlying an EMT of murine mammary epithelial cells and breast cancer cells. This screen identified the MEK5-ERK5 axis as a critical player in TGFβ-mediated EMT. Suppression of MEK5-ERK5 signaling completely prevented the morphological and molecular changes occurring during a TGFβ-induced EMT and, conversely, forced highly metastatic breast cancer cells into a differentiated epithelial state. Inhibition of MEK5-ERK5 signaling also repressed breast cancer cell migration and invasion and substantially reduced lung metastasis without affecting primary tumor growth. The results suggest that the MEK5-ERK5 signaling axis via activation of MEF2B and other transcription factors plays an important role in the induction and maintenance of breast cancer cell migration and invasion and thus represents an exploitable target for the pharmacological inhibition of cancer cell metastasis.
Collapse
Affiliation(s)
- Simona Pavan
- Department of Biomedicine, University of Basel, Basel, 4058, Switzerland.
| | | | - Maren Diepenbruck
- Department of Biomedicine, University of Basel, Basel, 4058, Switzerland
| | | | - Meera Saxena
- Department of Biomedicine, University of Basel, Basel, 4058, Switzerland
| | | |
Collapse
|
72
|
Siddharth J, Chakrabarti A, Pannérec A, Karaz S, Morin-Rivron D, Masoodi M, Feige JN, Parkinson SJ. Aging and sarcopenia associate with specific interactions between gut microbes, serum biomarkers and host physiology in rats. Aging (Albany NY) 2018; 9:1698-1720. [PMID: 28783713 PMCID: PMC5559170 DOI: 10.18632/aging.101262] [Citation(s) in RCA: 84] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Accepted: 07/12/2017] [Indexed: 12/24/2022]
Abstract
The microbiome has been demonstrated to play an integral role in the maintenance of many aspects of health that are also associated with aging. In order to identify areas of potential exploration and intervention, we simultaneously characterized age-related alterations in gut microbiome, muscle physiology and serum proteomic and lipidomic profiles in aged rats to define an integrated signature of the aging phenotype. We demonstrate that aging skews the composition of the gut microbiome, in particular by altering the Sutterella to Barneseilla ratio, and alters the metabolic potential of intestinal bacteria. Age-related changes of the gut microbiome were associated with the physiological decline of musculoskeletal function, and with molecular markers of nutrient processing/availability, and inflammatory/immune status in aged versus adult rats. Altogether, our study highlights that aging leads to a complex interplay between the microbiome and host physiology, and provides candidate microbial species to target physical and metabolic decline during aging by modulating gut microbial ecology.
Collapse
Affiliation(s)
- Jay Siddharth
- Nestlé Institute of Health Sciences SA, EPFL Innovation Park, Lausanne 1015, Switzerland
| | - Anirikh Chakrabarti
- Nestlé Institute of Health Sciences SA, EPFL Innovation Park, Lausanne 1015, Switzerland
| | - Alice Pannérec
- Nestlé Institute of Health Sciences SA, EPFL Innovation Park, Lausanne 1015, Switzerland
| | - Sonia Karaz
- Nestlé Institute of Health Sciences SA, EPFL Innovation Park, Lausanne 1015, Switzerland
| | - Delphine Morin-Rivron
- Nestlé Institute of Health Sciences SA, EPFL Innovation Park, Lausanne 1015, Switzerland
| | - Mojgan Masoodi
- Nestlé Institute of Health Sciences SA, EPFL Innovation Park, Lausanne 1015, Switzerland
| | - Jerome N Feige
- Nestlé Institute of Health Sciences SA, EPFL Innovation Park, Lausanne 1015, Switzerland
| | - Scott James Parkinson
- Nestlé Institute of Health Sciences SA, EPFL Innovation Park, Lausanne 1015, Switzerland
| |
Collapse
|
73
|
Nguyen EV, Centenera MM, Moldovan M, Das R, Irani S, Vincent AD, Chan H, Horvath LG, Lynn DJ, Daly RJ, Butler LM. Identification of Novel Response and Predictive Biomarkers to Hsp90 Inhibitors Through Proteomic Profiling of Patient-derived Prostate Tumor Explants. Mol Cell Proteomics 2018; 17:1470-1486. [PMID: 29632047 DOI: 10.1074/mcp.ra118.000633] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Revised: 03/26/2018] [Indexed: 12/16/2022] Open
Abstract
Inhibition of the heat shock protein 90 (Hsp90) chaperone is a promising therapeutic strategy to target expression of the androgen receptor (AR) and other oncogenic drivers in prostate cancer cells. However, identification of clinically-relevant responses and predictive biomarkers is essential to maximize efficacy and treatment personalization. Here, we combined mass spectrometry (MS)-based proteomic analyses with a unique patient-derived explant (PDE) model that retains the complex microenvironment of primary prostate tumors. Independent discovery and validation cohorts of PDEs (n = 16 and 30, respectively) were cultured in the absence or presence of Hsp90 inhibitors AUY922 or 17-AAG. PDEs were analyzed by LC-MS/MS with a hyper-reaction monitoring data independent acquisition (HRM-DIA) workflow, and differentially expressed proteins identified using repeated measure analysis of variance (ANOVA; raw p value <0.01). Using gene set enrichment, we found striking conservation of the most significantly AUY922-altered gene pathways between the discovery and validation cohorts, indicating that our experimental and analysis workflows were robust. Eight proteins were selectively altered across both cohorts by the most potent inhibitor, AUY922, including TIMP1, SERPINA3 and CYP51A (adjusted p < 0.01). The AUY922-mediated decrease in secretory TIMP1 was validated by ELISA of the PDE culture medium. We next exploited the heterogeneous response of PDEs to 17-AAG in order to detect predictive biomarkers of response and identified PCBP3 as a marker with increased expression in PDEs that had no response or increased in proliferation. Also, 17-AAG treatment led to increased expression of DNAJA1 in PDEs that exhibited a cytostatic response, revealing potential drug resistance mechanisms. This selective regulation of DNAJA1 was validated by Western blot analysis. Our study establishes "proof-of-principle" that proteomic profiling of drug-treated PDEs represents an effective and clinically-relevant strategy for identification of biomarkers that associate with certain tumor-specific responses.
Collapse
Affiliation(s)
- Elizabeth V Nguyen
- From the ‡Cancer Program, Biomedicine Discovery Institute, Monash University, Clayton, Victoria 3800, Australia.,§Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria 3800, Australia
| | - Margaret M Centenera
- ¶Adelaide Medical School and Freemasons Foundation Centre for Men's Health, University of Adelaide, Adelaide, South Australia 5005, Australia.,‖South Australian Health and Medical Research Institute, Adelaide, South Australia 5000, Australia
| | - Max Moldovan
- ‖South Australian Health and Medical Research Institute, Adelaide, South Australia 5000, Australia
| | - Rajdeep Das
- ¶Adelaide Medical School and Freemasons Foundation Centre for Men's Health, University of Adelaide, Adelaide, South Australia 5005, Australia
| | - Swati Irani
- ¶Adelaide Medical School and Freemasons Foundation Centre for Men's Health, University of Adelaide, Adelaide, South Australia 5005, Australia.,‖South Australian Health and Medical Research Institute, Adelaide, South Australia 5000, Australia
| | - Andrew D Vincent
- ¶Adelaide Medical School and Freemasons Foundation Centre for Men's Health, University of Adelaide, Adelaide, South Australia 5005, Australia
| | - Howard Chan
- From the ‡Cancer Program, Biomedicine Discovery Institute, Monash University, Clayton, Victoria 3800, Australia.,§Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria 3800, Australia
| | - Lisa G Horvath
- **Cancer Division, The Kinghorn Cancer Centre/Garvan Institute of Medical Research, Darlinghurst, New South Wales 2010, Australia.,‡‡Royal Prince Alfred Hospital, Camperdown, New South Wales 2050, Australia.,§§Department of Medical Oncology, Chris O'Brien Lifehouse, Camperdown, New South Wales 2050, Australia
| | - David J Lynn
- ‖South Australian Health and Medical Research Institute, Adelaide, South Australia 5000, Australia.,¶¶School of Medicine, Flinders University, Bedford Park, SA 5042, Australia
| | - Roger J Daly
- From the ‡Cancer Program, Biomedicine Discovery Institute, Monash University, Clayton, Victoria 3800, Australia; .,§Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria 3800, Australia
| | - Lisa M Butler
- ¶Adelaide Medical School and Freemasons Foundation Centre for Men's Health, University of Adelaide, Adelaide, South Australia 5005, Australia.,‖South Australian Health and Medical Research Institute, Adelaide, South Australia 5000, Australia
| |
Collapse
|
74
|
Sack LM, Davoli T, Li MZ, Li Y, Xu Q, Naxerova K, Wooten EC, Bernardi RJ, Martin TD, Chen T, Leng Y, Liang AC, Scorsone KA, Westbrook TF, Wong KK, Elledge SJ. Profound Tissue Specificity in Proliferation Control Underlies Cancer Drivers and Aneuploidy Patterns. Cell 2018; 173:499-514.e23. [PMID: 29576454 PMCID: PMC6643283 DOI: 10.1016/j.cell.2018.02.037] [Citation(s) in RCA: 129] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Revised: 11/10/2017] [Accepted: 02/15/2018] [Indexed: 02/06/2023]
Abstract
Genomics has provided a detailed structural description of the cancer genome. Identifying oncogenic drivers that work primarily through dosage changes is a current challenge. Unrestrained proliferation is a critical hallmark of cancer. We constructed modular, barcoded libraries of human open reading frames (ORFs) and performed screens for proliferation regulators in multiple cell types. Approximately 10% of genes regulate proliferation, with most performing in an unexpectedly highly tissue-specific manner. Proliferation drivers in a given cell type showed specific enrichment in somatic copy number changes (SCNAs) from cognate tumors and helped predict aneuploidy patterns in those tumors, implying that tissue-type-specific genetic network architectures underlie SCNA and driver selection in different cancers. In vivo screening confirmed these results. We report a substantial contribution to the catalog of SCNA-associated cancer drivers, identifying 147 amplified and 107 deleted genes as potential drivers, and derive insights about the genetic network architecture of aneuploidy in tumors.
Collapse
Affiliation(s)
- Laura Magill Sack
- Division of Genetics, Brigham and Women's Hospital, Howard Hughes Medical Institute, Department of Genetics, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Teresa Davoli
- Division of Genetics, Brigham and Women's Hospital, Howard Hughes Medical Institute, Department of Genetics, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Mamie Z Li
- Division of Genetics, Brigham and Women's Hospital, Howard Hughes Medical Institute, Department of Genetics, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Yuyang Li
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA; Shandong Provincial Hospital affiliated to Shandong University, Jinan 250021, China
| | - Qikai Xu
- Division of Genetics, Brigham and Women's Hospital, Howard Hughes Medical Institute, Department of Genetics, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Kamila Naxerova
- Division of Genetics, Brigham and Women's Hospital, Howard Hughes Medical Institute, Department of Genetics, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Eric C Wooten
- Division of Genetics, Brigham and Women's Hospital, Howard Hughes Medical Institute, Department of Genetics, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Ronald J Bernardi
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Department of Molecular and Human Genetics, Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Timothy D Martin
- Division of Genetics, Brigham and Women's Hospital, Howard Hughes Medical Institute, Department of Genetics, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Ting Chen
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA; Laura and Isaac Perlmutter Cancer Center, New York University Langone Medical Center, New York, NY, USA
| | - Yumei Leng
- Division of Genetics, Brigham and Women's Hospital, Howard Hughes Medical Institute, Department of Genetics, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Anthony C Liang
- Division of Genetics, Brigham and Women's Hospital, Howard Hughes Medical Institute, Department of Genetics, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Kathleen A Scorsone
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Department of Molecular and Human Genetics, Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Thomas F Westbrook
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Department of Molecular and Human Genetics, Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Kwok-Kin Wong
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA; Laura and Isaac Perlmutter Cancer Center, New York University Langone Medical Center, New York, NY, USA
| | - Stephen J Elledge
- Division of Genetics, Brigham and Women's Hospital, Howard Hughes Medical Institute, Department of Genetics, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA.
| |
Collapse
|
75
|
Moradzadeh K, Moein S, Nickaeen N, Gheisari Y. Analysis of time-course microarray data: Comparison of common tools. Genomics 2018; 111:636-641. [PMID: 29614346 DOI: 10.1016/j.ygeno.2018.03.021] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2017] [Revised: 03/27/2018] [Accepted: 03/30/2018] [Indexed: 12/11/2022]
Abstract
High-throughput time-series data have a special value for studying the dynamism of biological systems. However, the interpretation of such complex data can be challenging. The aim of this study was to compare common algorithms recently developed for the detection of differentially expressed genes in time-course microarray data. Using different measures such as sensitivity, specificity, predictive values, and related signaling pathways, we found that limma, timecourse, and gprege have reasonably good performance for the analysis of datasets in which only test group is followed over time. However, limma has the additional advantage of being able to report significance cut off, making it a more practical tool. In addition, limma and TTCA can be satisfactorily used for datasets with time-series data for all experimental groups. These findings may assist investigators to select appropriate tools for the detection of differentially expressed genes as an initial step in the interpretation of time-course big data.
Collapse
Affiliation(s)
- Kobra Moradzadeh
- Department of Genetics and Molecular Biology, Isfahan University of Medical Sciences, Isfahan, Iran; Student Research Committee, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - Shiva Moein
- Department of Genetics and Molecular Biology, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - Niloofar Nickaeen
- Department of Electrical and Computer Engineering, Isfahan University of Technology, Isfahan, Iran.
| | - Yousof Gheisari
- Department of Genetics and Molecular Biology, Isfahan University of Medical Sciences, Isfahan, Iran; Regenerative Medicine Lab, Isfahan Kidney Diseases Research Center, Isfahan University of Medical Sciences, Isfahan, Iran.
| |
Collapse
|
76
|
Racz R, Soldatos TG, Jackson D, Burkhart K. Association Between Serotonin Syndrome and Second-Generation Antipsychotics via Pharmacological Target-Adverse Event Analysis. Clin Transl Sci 2018; 11:322-329. [PMID: 29575568 PMCID: PMC5944571 DOI: 10.1111/cts.12543] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Accepted: 01/21/2018] [Indexed: 12/25/2022] Open
Abstract
Case reports suggest an association between second‐generation antipsychotics (SGAs) and serotonin syndrome (SS). The US Food and Drug Administration (FDA) Adverse Event Reporting System (FAERS) was analyzed to generate hypotheses about how SGAs may interact with pharmacological targets associated with SS. FAERS was integrated with additional sources to link information about adverse events with drugs and targets. Using Proportional Reporting Ratios, we identified signals that were further investigated with the literature to evaluate mechanistic hypotheses formed from the integrated FAERS data. Analysis revealed common pharmacological targets perturbed in both SGA and SS cases, indicating that SGAs may induce SS. The literature also supported 5‐HT2A antagonism and 5‐HT1A agonism as common mechanisms that may explain the SGA‐SS association. Additionally, integrated FAERS data mining and case studies suggest that interactions between SGAs and other serotonergic agents may increase the risk for SS. Computational analysis can provide additional insights into the mechanisms underlying the relationship between SGAs and SS.
Collapse
Affiliation(s)
- Rebecca Racz
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, USA
| | | | | | - Keith Burkhart
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, USA
| |
Collapse
|
77
|
Lou Y, Jiang H, Cui Z, Wang X, Wang L, Han Y. Gene microarray analysis of lncRNA and mRNA expression profiles in patients with high‑grade ovarian serous cancer. Int J Mol Med 2018; 42:91-104. [PMID: 29577163 PMCID: PMC5979786 DOI: 10.3892/ijmm.2018.3588] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Accepted: 03/16/2018] [Indexed: 12/15/2022] Open
Abstract
High-grade ovarian serous cancer is known for its high rates of invasion and metastasis, and resultant high mortality rate. Therefore, research concerning biomarkers and underlying molecular mechanisms of high-grade ovarian serous cancer progression and prognosis are urgently required. Long non-coding RNAs (lncRNAs) have been the subject of an increasing number of studies, and certain lncRNAs have been demonstrated to serve an important function in the development and progression of various cancers, including HOX transcript antisense RNA, competing endogenous lncRNA 2 for microRNA let-7b, urothelial cancer associated 1, and H19, imprinted maternally expressed transcript (non-protein coding). However, few studies have investigated the differential expression of lncRNAs in high-grade ovarian serous cancer. In the present study, differences in lncRNA and mRNA expression profiles between high-grade ovarian serous cancer tissue samples and healthy fallopian tube tissue samples were investigated using microarray analysis, and the differential expression of lncRNAs and mRNAs was confirmed by reverse transcription-quantitative polymerase chain reaction (RT-qPCR). Then, five abnormally expressed lncRNAs were selected, and the associations between these lncRNAs and ovarian cancer clinicopathological parameters were examined using RT-qPCR. The expression profiles of certain lncRNAs and mRNAs were confirmed to be altered between high-grade ovarian serous cancer tissues and healthy fallopian tube tissues. Furthermore, the expression levels of selected lncRNAs were associated with International Federation of Gynecology and Obstetrics stage and lymph node metastasis. These lncRNAs and mRNAs may therefore be involved in the pathogenesis of high-grade ovarian serous cancer. The results of the present study provide an experimental foundation for further exploration of the value of these lncRNAs and mRNAs in the early diagnosis and treatment of high-grade ovarian serous cancer.
Collapse
Affiliation(s)
- Yanhui Lou
- Department of Gynecology, Affiliated Hospital of Qingdao University, Qingdao, Shandong 266100, P.R. China
| | - Huanhuan Jiang
- Department of Gynecology, Affiliated Hospital of Qingdao University, Qingdao, Shandong 266100, P.R. China
| | - Zhumei Cui
- Department of Gynecology, Affiliated Hospital of Qingdao University, Qingdao, Shandong 266100, P.R. China
| | - Xiangyu Wang
- Department of Gynecology, Affiliated Hospital of Qingdao University, Qingdao, Shandong 266100, P.R. China
| | - Lingzhi Wang
- Department of Gynecology, Affiliated Hospital of Qingdao University, Qingdao, Shandong 266100, P.R. China
| | - Yi Han
- Department of Gynecology, Affiliated Hospital of Qingdao University, Qingdao, Shandong 266100, P.R. China
| |
Collapse
|
78
|
Dickinson JM, D'Lugos AC, Naymik MA, Siniard AL, Wolfe AJ, Curtis DR, Huentelman MJ, Carroll CC. Transcriptome response of human skeletal muscle to divergent exercise stimuli. J Appl Physiol (1985) 2018. [PMID: 29543133 DOI: 10.1152/japplphysiol.00014.2018] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Aerobic (AE) and resistance exercise (RE) elicit unique adaptations in skeletal muscle that have distinct implications for health and performance. The purpose of this study was to identify the unique transcriptome response of skeletal muscle to acute AE and RE. In a counterbalanced, crossover design, six healthy, recreationally active young men (27 ± 3 yr) completed acute AE (40 min of cycling, ∼70% maximal HR) and RE [8 sets, 10 reps, ∼65% 1-repetition maximum (1RM)], separated by ∼1 wk. Muscle biopsies (vastus lateralis) were obtained before and at 1 and 4 h postexercise. Whole transcriptome RNA sequencing (HiSeq2500; Illumina) was performed on cDNA synthesized from skeletal muscle RNA. Sequencing data were analyzed using HTSeq, and differential gene expression was identified using DESeq2 [adjusted P value (FDR) <0.05, >1.5-fold change from preexercise]. RE resulted in a greater number of differentially expressed genes at 1 (67 vs. 48) and 4 h (523 vs. 221) compared with AE. We identified 348 genes that were differentially expressed only following RE, whereas 48 genes were differentially expressed only following AE. Gene clustering indicated that AE targeted functions related to zinc interaction, angiogenesis, and ubiquitination, whereas RE targeted functions related to transcription regulation, cytokine activity, cell adhesion, kinase activity, and the phosphatidylinositol 3-kinase (PI3K)/Akt pathway. ESRRG and TNFSRF12A were identified as potential targets related to the specific response of skeletal muscle to AE and RE, respectively. These data describe the early postexercise transcriptome response of skeletal muscle to acute AE and RE and further highlight that different forms of exercise stimulate unique molecular activity in skeletal muscle. NEW & NOTEWORTHY Whole transcriptome RNA sequencing was used to determine the early postexercise transcriptome response of skeletal muscle to acute aerobic (AE) and resistance exercise (RE) in untrained individuals. Although a number of shared genes were stimulated following both AE and RE, several genes were uniquely responsive to each exercise mode. These findings support the need for future research focused to better identify the role of exercise mode as it relates to targeting specific cellular skeletal muscle abnormalities.
Collapse
Affiliation(s)
- Jared M Dickinson
- School of Nutrition and Health Promotion, Healthy Lifestyles Research Center, Exercise Science and Health Promotion, Arizona State University , Phoenix, Arizona
| | - Andrew C D'Lugos
- School of Nutrition and Health Promotion, Healthy Lifestyles Research Center, Exercise Science and Health Promotion, Arizona State University , Phoenix, Arizona
| | - Marcus A Naymik
- Translational Genomics Research Institute , Phoenix, Arizona
| | | | - Amanda J Wolfe
- Translational Genomics Research Institute , Phoenix, Arizona
| | | | | | - Chad C Carroll
- Midwestern University , Glendale, Arizona.,Department of Health and Kinesiology, Purdue University , West Lafayette, Indiana
| |
Collapse
|
79
|
Bobisse S, Genolet R, Roberti A, Tanyi JL, Racle J, Stevenson BJ, Iseli C, Michel A, Le Bitoux MA, Guillaume P, Schmidt J, Bianchi V, Dangaj D, Fenwick C, Derré L, Xenarios I, Michielin O, Romero P, Monos DS, Zoete V, Gfeller D, Kandalaft LE, Coukos G, Harari A. Sensitive and frequent identification of high avidity neo-epitope specific CD8 + T cells in immunotherapy-naive ovarian cancer. Nat Commun 2018; 9:1092. [PMID: 29545564 PMCID: PMC5854609 DOI: 10.1038/s41467-018-03301-0] [Citation(s) in RCA: 110] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Accepted: 02/05/2018] [Indexed: 12/23/2022] Open
Abstract
Immunotherapy directed against private tumor neo-antigens derived from non-synonymous somatic mutations is a promising strategy of personalized cancer immunotherapy. However, feasibility in low mutational load tumor types remains unknown. Comprehensive and deep analysis of circulating and tumor-infiltrating lymphocytes (TILs) for neo-epitope specific CD8+ T cells has allowed prompt identification of oligoclonal and polyfunctional such cells from most immunotherapy-naive patients with advanced epithelial ovarian cancer studied. Neo-epitope recognition is discordant between circulating T cells and TILs, and is more likely to be found among TILs, which display higher functional avidity and unique TCRs with higher predicted affinity than their blood counterparts. Our results imply that identification of neo-epitope specific CD8+ T cells is achievable even in tumors with relatively low number of somatic mutations, and neo-epitope validation in TILs extends opportunities for mutanome-based personalized immunotherapies to such tumors.
Collapse
Affiliation(s)
- Sara Bobisse
- Department of Oncology, Lausanne University Hospital, Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, CH-1066, Switzerland
| | - Raphael Genolet
- Department of Oncology, Lausanne University Hospital, Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, CH-1066, Switzerland
| | - Annalisa Roberti
- Ovarian Cancer Research Center, Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Janos L Tanyi
- Ovarian Cancer Research Center, Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Julien Racle
- Department of Oncology, Lausanne University Hospital, Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, CH-1066, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, CH-1015, Switzerland
| | | | - Christian Iseli
- Swiss Institute of Bioinformatics, Lausanne, CH-1015, Switzerland
| | - Alexandra Michel
- Department of Oncology, Lausanne University Hospital, Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, CH-1066, Switzerland
| | - Marie-Aude Le Bitoux
- Department of Oncology, Lausanne University Hospital, Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, CH-1066, Switzerland
| | - Philippe Guillaume
- Department of Oncology, Lausanne University Hospital, Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, CH-1066, Switzerland
| | - Julien Schmidt
- Department of Oncology, Lausanne University Hospital, Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, CH-1066, Switzerland
| | - Valentina Bianchi
- Department of Oncology, Lausanne University Hospital, Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, CH-1066, Switzerland
| | - Denarda Dangaj
- Department of Oncology, Lausanne University Hospital, Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, CH-1066, Switzerland
| | - Craig Fenwick
- Department of Medicine, Division of Immunology and Allergy, Lausanne University Hospital, Lausanne, CH-1066, Switzerland
| | - Laurent Derré
- Urology Research Unit, Lausanne University Hospital, Lausanne, CH-1011, Switzerland
| | - Ioannis Xenarios
- Swiss Institute of Bioinformatics, Lausanne, CH-1015, Switzerland
| | - Olivier Michielin
- Department of Oncology, Lausanne University Hospital, Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, CH-1066, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, CH-1015, Switzerland
| | - Pedro Romero
- Department of Oncology, Lausanne University Hospital, Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, CH-1066, Switzerland
| | - Dimitri S Monos
- Department of Pathology and Laboratory Medicine, Immunogenetics Laboratory, The Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Vincent Zoete
- Department of Oncology, Lausanne University Hospital, Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, CH-1066, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, CH-1015, Switzerland
| | - David Gfeller
- Department of Oncology, Lausanne University Hospital, Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, CH-1066, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, CH-1015, Switzerland
| | - Lana E Kandalaft
- Department of Oncology, Lausanne University Hospital, Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, CH-1066, Switzerland
- Ovarian Cancer Research Center, Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - George Coukos
- Department of Oncology, Lausanne University Hospital, Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, CH-1066, Switzerland.
| | - Alexandre Harari
- Department of Oncology, Lausanne University Hospital, Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, CH-1066, Switzerland.
| |
Collapse
|
80
|
Zhang B, Hu S, Baskin E, Patt A, Siddiqui JK, Mathé EA. RaMP: A Comprehensive Relational Database of Metabolomics Pathways for Pathway Enrichment Analysis of Genes and Metabolites. Metabolites 2018; 8:E16. [PMID: 29470400 PMCID: PMC5876005 DOI: 10.3390/metabo8010016] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Revised: 02/14/2018] [Accepted: 02/16/2018] [Indexed: 01/04/2023] Open
Abstract
The value of metabolomics in translational research is undeniable, and metabolomics data are increasingly generated in large cohorts. The functional interpretation of disease-associated metabolites though is difficult, and the biological mechanisms that underlie cell type or disease-specific metabolomics profiles are oftentimes unknown. To help fully exploit metabolomics data and to aid in its interpretation, analysis of metabolomics data with other complementary omics data, including transcriptomics, is helpful. To facilitate such analyses at a pathway level, we have developed RaMP (Relational database of Metabolomics Pathways), which combines biological pathways from the Kyoto Encyclopedia of Genes and Genomes (KEGG), Reactome, WikiPathways, and the Human Metabolome DataBase (HMDB). To the best of our knowledge, an off-the-shelf, public database that maps genes and metabolites to biochemical/disease pathways and can readily be integrated into other existing software is currently lacking. For consistent and comprehensive analysis, RaMP enables batch and complex queries (e.g., list all metabolites involved in glycolysis and lung cancer), can readily be integrated into pathway analysis tools, and supports pathway overrepresentation analysis given a list of genes and/or metabolites of interest. For usability, we have developed a RaMP R package (https://github.com/Mathelab/RaMP-DB), including a user-friendly RShiny web application, that supports basic simple and batch queries, pathway overrepresentation analysis given a list of genes or metabolites of interest, and network visualization of gene-metabolite relationships. The package also includes the raw database file (mysql dump), thereby providing a stand-alone downloadable framework for public use and integration with other tools. In addition, the Python code needed to recreate the database on another system is also publicly available (https://github.com/Mathelab/RaMP-BackEnd). Updates for databases in RaMP will be checked multiple times a year and RaMP will be updated accordingly.
Collapse
Affiliation(s)
- Bofei Zhang
- Department of Biomedical Informatics, College of Medicine, The Ohio State University, Columbus, OH 43210, USA.
| | - Senyang Hu
- Department of Biomedical Informatics, College of Medicine, The Ohio State University, Columbus, OH 43210, USA.
| | - Elizabeth Baskin
- Department of Biomedical Informatics, College of Medicine, The Ohio State University, Columbus, OH 43210, USA.
| | - Andrew Patt
- Department of Biomedical Informatics, College of Medicine, The Ohio State University, Columbus, OH 43210, USA.
- Biomedical Engineering Graduate Program, The Ohio State University, Columbus, OH 43210, USA.
| | - Jalal K Siddiqui
- Department of Biomedical Informatics, College of Medicine, The Ohio State University, Columbus, OH 43210, USA.
| | - Ewy A Mathé
- Department of Biomedical Informatics, College of Medicine, The Ohio State University, Columbus, OH 43210, USA.
| |
Collapse
|
81
|
Tanno B, Babini G, Leonardi S, Giardullo P, De Stefano I, Pasquali E, Ottolenghi A, Atkinson MJ, Saran A, Mancuso M. Ex vivo miRNome analysis in Ptch1+/- cerebellum granule cells reveals a subset of miRNAs involved in radiation-induced medulloblastoma. Oncotarget 2018; 7:68253-68269. [PMID: 27626168 PMCID: PMC5356552 DOI: 10.18632/oncotarget.11938] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Accepted: 09/05/2016] [Indexed: 12/15/2022] Open
Abstract
It has historically been accepted that incorrectly repaired DNA double strand breaks (DSBs) are the principal lesions of importance regarding mutagenesis, and long-term biological effects associated with ionizing radiation. However, radiation may also cause dysregulation of epigenetic processes that can lead to altered gene function and malignant transformation, and epigenetic alterations are important causes of miRNAs dysregulation in cancer. Patched1 heterozygous (Ptch1+/−) mice, characterized by aberrant activation of the Sonic hedgehog (Shh) signaling pathway, are a well-known murine model of spontaneous and radiation-induced medulloblastoma (MB), a common pediatric brain tumor originating from neural granule cell progenitors (GCPs). The high sensitivity of neonatal Ptch1+/− mice to radiogenic MB is dependent on deregulation of the Ptch1 gene function. Ptch1 activates a growth and differentiation programme that is a strong candidate for regulation through the non-coding genome. Therefore we carried out miRNA next generation sequencing in ex vivo irradiated and control GCPs, isolated and purified from cerebella of neonatal WT and Ptch1+/− mice. We identified a subset of miRNAs, namely let-7 family and miR-17∼92 cluster members, whose expression is altered in GCPs by radiation alone, or by synergistic interaction of radiation with Shh-deregulation. The same miRNAs were further validated in spontaneous and radiation-induced MBs from Ptch1+/− mice, confirming persistent deregulation of these miRNAs in the pathogenesis of MB. Our results support the hypothesis that miRNAs dysregulation is associated with radiosensitivity of GCPs and their neoplastic transformation in vivo.
Collapse
Affiliation(s)
- Barbara Tanno
- Laboratory of Biomedical Technologies, Agenzia Nazionale per le Nuove Tecnologie, l'Energia e lo Sviluppo Economico Sostenibile (ENEA), Rome, Italy
| | | | - Simona Leonardi
- Laboratory of Biomedical Technologies, Agenzia Nazionale per le Nuove Tecnologie, l'Energia e lo Sviluppo Economico Sostenibile (ENEA), Rome, Italy
| | - Paola Giardullo
- Department of Radiation Physics, Guglielmo Marconi University, Rome, Italy.,Department of Sciences, Roma Tre University, Rome, Italy
| | - Ilaria De Stefano
- Department of Radiation Physics, Guglielmo Marconi University, Rome, Italy
| | - Emanuela Pasquali
- Laboratory of Biomedical Technologies, Agenzia Nazionale per le Nuove Tecnologie, l'Energia e lo Sviluppo Economico Sostenibile (ENEA), Rome, Italy
| | | | - Michael J Atkinson
- Helmholtz Zentrum München, German Research Center for Environmental Health, Institute of Radiation Biology, Neuherberg, Germany
| | - Anna Saran
- Laboratory of Biomedical Technologies, Agenzia Nazionale per le Nuove Tecnologie, l'Energia e lo Sviluppo Economico Sostenibile (ENEA), Rome, Italy
| | - Mariateresa Mancuso
- Laboratory of Biomedical Technologies, Agenzia Nazionale per le Nuove Tecnologie, l'Energia e lo Sviluppo Economico Sostenibile (ENEA), Rome, Italy
| |
Collapse
|
82
|
Rondeau G, Abedinpour P, Chrastina A, Pelayo J, Borgstrom P, Welsh J. Differential gene expression induced by anti-cancer agent plumbagin is mediated by androgen receptor in prostate cancer cells. Sci Rep 2018; 8:2694. [PMID: 29426892 PMCID: PMC5807367 DOI: 10.1038/s41598-018-20451-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Accepted: 01/15/2018] [Indexed: 12/24/2022] Open
Abstract
Treatment of mice harboring PTEN-P2 tumors in the prostate or on prostate tissue in vivo with 5-hydroxy-2-methyl-1,4-naphthoquinone, also known as plumbagin, results in tumor regression in castrated mice, but not in intact mice. This suggested that dihydrotestosterone (DHT) production in the testes may prevent cell death due to plumbagin treatment, but the underlying mechanism is not understood. We performed RNA-seq analysis on cells treated with combinations of plumbagin and DHT, and analyzed differential gene expression, to gain insight into the interactions between androgen and plumbgin. DHT and plumbagin synergize to alter the expression of many genes that are not differentially regulated by either single agent when used alone. These experiments revealed that, for many genes, increases in mRNAs caused by DHT are sharply down-regulated by plumbagin, and that many transcripts change in response to plumbagin in a DHT-dependent manner. This suggests that androgen receptor mediates some of the effects of plumbagin on gene expression.
Collapse
Affiliation(s)
- Gaelle Rondeau
- Vaccine Research Institute of San Diego, 3030 Bunker Hill Street, Suite 200, San Diego, CA, 92109, USA
| | - Parisa Abedinpour
- Vaccine Research Institute of San Diego, 3030 Bunker Hill Street, Suite 200, San Diego, CA, 92109, USA
| | - Adrian Chrastina
- Vaccine Research Institute of San Diego, 3030 Bunker Hill Street, Suite 200, San Diego, CA, 92109, USA
| | - Jennifer Pelayo
- Vaccine Research Institute of San Diego, 3030 Bunker Hill Street, Suite 200, San Diego, CA, 92109, USA
| | - Per Borgstrom
- Vaccine Research Institute of San Diego, 3030 Bunker Hill Street, Suite 200, San Diego, CA, 92109, USA
- Pellficure Pharmaceuticals, Inc., 2325 Camino del Collado, La Jolla, CA, 92037, USA
| | - John Welsh
- Vaccine Research Institute of San Diego, 3030 Bunker Hill Street, Suite 200, San Diego, CA, 92109, USA.
| |
Collapse
|
83
|
Chung SS, Eng WS, Hu W, Khalaj M, Garrett-Bakelman FE, Tavakkoli M, Levine RL, Carroll M, Klimek VM, Melnick AM, Park CY. CD99 is a therapeutic target on disease stem cells in myeloid malignancies. Sci Transl Med 2018; 9:9/374/eaaj2025. [PMID: 28123069 DOI: 10.1126/scitranslmed.aaj2025] [Citation(s) in RCA: 105] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Revised: 09/12/2016] [Accepted: 11/22/2016] [Indexed: 12/16/2022]
Abstract
Acute myeloid leukemia (AML) and the myelodysplastic syndromes (MDS) are initiated and sustained by self-renewing malignant stem cells; thus, eradication of AML and MDS stem cells is required for cure. We identified CD99 as a cell surface protein frequently overexpressed on AML and MDS stem cells. Expression of CD99 allows for prospective separation of leukemic stem cells (LSCs) from functionally normal hematopoietic stem cells in AML, and high CD99 expression on AML blasts enriches for functional LSCs as demonstrated by limiting dilution xenotransplant studies. Monoclonal antibodies (mAbs) targeting CD99 induce the death of AML and MDS cells in a SARC family kinase-dependent manner in the absence of immune effector cells or complement, and anti-CD99 mAbs exhibit antileukemic activity in AML xenografts. These data establish CD99 as a marker of AML and MDS stem cells, as well as a promising therapeutic target in these disorders.
Collapse
Affiliation(s)
- Stephen S Chung
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.,Leukemia Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - William S Eng
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Wenhuo Hu
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Mona Khalaj
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.,Weill Graduate School of Medical Sciences, Cornell University, New York, NY 10065, USA
| | - Francine E Garrett-Bakelman
- Division of Hematology and Medical Oncology, Department of Medicine, Weill Cornell School of Medicine, New York, NY 10065, USA
| | - Montreh Tavakkoli
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Ross L Levine
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.,Leukemia Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Martin Carroll
- Division of Hematology and Oncology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Virginia M Klimek
- Leukemia Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Ari M Melnick
- Division of Hematology and Medical Oncology, Department of Medicine, Weill Cornell School of Medicine, New York, NY 10065, USA
| | - Christopher Y Park
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA. .,Departments of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| |
Collapse
|
84
|
Rask-Andersen M, Martinsson D, Ahsan M, Enroth S, Ek WE, Gyllensten U, Johansson Å. Epigenome-wide association study reveals differential DNA methylation in individuals with a history of myocardial infarction. Hum Mol Genet 2018; 25:4739-4748. [PMID: 28172975 DOI: 10.1093/hmg/ddw302] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Revised: 08/24/2016] [Accepted: 08/26/2016] [Indexed: 12/24/2022] Open
Abstract
Cardiovascular diseases (CVDs) are the leading causes of death worldwide and represent a substantial economic burden on public health care systems. Epigenetic markers have potential as diagnostic markers before clinical symptoms have emerged, and as prognostic markers to inform the choice of clinical intervention. In this study, we performed an epigenome-wide association study (EWAS) for CVDs, to identify disease-specific alterations in DNA methylation. CpG methylation in blood samples from the northern Sweden population health study (NSPHS) (n = 729) was assayed on the Illumina Infinium HumanMethylation450 BeadChip. Individuals with a history of a CVD were identified in the cohort. It included individuals with hypertension (N = 147), myocardial infarction (MI) (N = 48), stroke (N = 27), thrombosis (N = 22) and cardiac arrhythmia (N = 5). Differential DNA methylation was observed at 211 CpG-sites in individuals with a history of MI (q <0.05). These sites represent 196 genes, of which 42 have been described in the scientific literature to be related to cardiac function, cardiovascular disease, cardiogenesis and recovery after ischemic injury. We have shown that individuals with a history of MI have a deviating pattern of DNA methylation at many genomic loci of which a large fraction has previously been linked to CVD. Our results highlight genes that might be important in the pathogenesis of MI or in recovery. In addition, the sites pointed out in this study can serve as candidates for further evaluation as potential biomarkers for MI.
Collapse
Affiliation(s)
- Mathias Rask-Andersen
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - David Martinsson
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Muhammad Ahsan
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Stefan Enroth
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Weronica E Ek
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Ulf Gyllensten
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Åsa Johansson
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| |
Collapse
|
85
|
Allen H, Shraga-Heled N, Blumenfeld M, Dego-Ashto T, Fuchs-Telem D, Gilert A, Aberman Z, Ofir R. Human Placental-Derived Adherent Stromal Cells Co-Induced with TNF-α and IFN-γ Inhibit Triple-Negative Breast Cancer in Nude Mouse Xenograft Models. Sci Rep 2018; 8:670. [PMID: 29330447 PMCID: PMC5766494 DOI: 10.1038/s41598-017-18428-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Accepted: 12/11/2017] [Indexed: 12/26/2022] Open
Abstract
Culturing 3D-expanded human placental-derived adherent stromal cells (ASCs) in the presence of tumor necrosis factor-alpha (TNF-α) and interferon-gamma (IFN-γ) transiently upregulated the secretion of numerous anti-proliferative, anti-angiogenic and pro-inflammatory cytokines. In a 3D-spheroid screening assay, conditioned medium from these induced-ASCs inhibited proliferation of cancer cell lines, including triple-negative breast cancer (TNBC) lines. In vitro co-culture studies of induced-ASCs with MDA-MB-231 human breast carcinoma cells, a model representing TNBC, supports a mechanism involving immunomodulation and angiogenesis inhibition. In vivo studies in nude mice showed that intramuscular administration of induced-ASCs halted MDA-MB-231 cell proliferation, and inhibited tumor progression and vascularization. Thirty percent of treated mice experienced complete tumor remission. Murine serum concentrations of the tumor-supporting cytokines Interleukin-6 (IL-6), Vascular endothelial growth factor (VEGF) and Granulocyte-colony stimulating factor (G-CSF) were lowered to naïve levels. A somatic mutation analysis identified numerous genes which could be screened in patients to increase a positive therapeutic outcome. Taken together, these results show that targeted changes in the secretion profile of ASCs may improve their therapeutic potential.
Collapse
|
86
|
Blood plasma/IgG N-glycome biosignatures associated with major depressive disorder symptom severity and the antidepressant response. Sci Rep 2018; 8:179. [PMID: 29317657 PMCID: PMC5760622 DOI: 10.1038/s41598-017-17500-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Accepted: 11/27/2017] [Indexed: 12/14/2022] Open
Abstract
While N-linked glycosylation has been extensively studied in the context of inflammatory and metabolic disorders, its relationship with major depressive disorder (MDD) and antidepressant treatment response has not been investigated. In our exploratory study, we analysed N-glycan profiles in blood plasma samples collected from MDD patients (n = 18) and found gender-dependent correlations with severity of depressive symptoms prior to initiating antidepressant treatment. In addition, several N-glycosylation traits showed gender-dependent associations with clinical antidepressant response. Follow up proteomics analysis in peripheral blood mononuclear cells (PBMCs) collected from MDD patients (n = 20) identified baseline and post-antidepressant treatment pathway differences between responder and non-responder patients. Reactome data analysis further delineated potential biological reaction differences between responder and non-responder patients. Our preliminary results suggest that specific glycosylation traits are associated with depressive symptom severity and antidepressant response and may be of use as biomarkers.
Collapse
|
87
|
Cava C, Bertoli G, Colaprico A, Olsen C, Bontempi G, Castiglioni I. Integration of multiple networks and pathways identifies cancer driver genes in pan-cancer analysis. BMC Genomics 2018; 19:25. [PMID: 29304754 PMCID: PMC5756345 DOI: 10.1186/s12864-017-4423-x] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Accepted: 12/27/2017] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Modern high-throughput genomic technologies represent a comprehensive hallmark of molecular changes in pan-cancer studies. Although different cancer gene signatures have been revealed, the mechanism of tumourigenesis has yet to be completely understood. Pathways and networks are important tools to explain the role of genes in functional genomic studies. However, few methods consider the functional non-equal roles of genes in pathways and the complex gene-gene interactions in a network. RESULTS We present a novel method in pan-cancer analysis that identifies de-regulated genes with a functional role by integrating pathway and network data. A pan-cancer analysis of 7158 tumour/normal samples from 16 cancer types identified 895 genes with a central role in pathways and de-regulated in cancer. Comparing our approach with 15 current tools that identify cancer driver genes, we found that 35.6% of the 895 genes identified by our method have been found as cancer driver genes with at least 2/15 tools. Finally, we applied a machine learning algorithm on 16 independent GEO cancer datasets to validate the diagnostic role of cancer driver genes for each cancer. We obtained a list of the top-ten cancer driver genes for each cancer considered in this study. CONCLUSIONS Our analysis 1) confirmed that there are several known cancer driver genes in common among different types of cancer, 2) highlighted that cancer driver genes are able to regulate crucial pathways.
Collapse
Affiliation(s)
- Claudia Cava
- Institute of Molecular Bioimaging and Physiology, National Research Council (IBFM-CNR), Via F.Cervi 93, 20090 Milan, Segrate-Milan Italy
| | - Gloria Bertoli
- Institute of Molecular Bioimaging and Physiology, National Research Council (IBFM-CNR), Via F.Cervi 93, 20090 Milan, Segrate-Milan Italy
| | - Antonio Colaprico
- Interuniversity Institute of Bioinformatics in Brussels (IB)2, 1050 Brussels, Belgium
- Machine Learning Group (MLG), Department d’Informatique, Universite libre de Bruxelles (ULB), 1050 Brussels, Belgium
| | - Catharina Olsen
- Interuniversity Institute of Bioinformatics in Brussels (IB)2, 1050 Brussels, Belgium
- Machine Learning Group (MLG), Department d’Informatique, Universite libre de Bruxelles (ULB), 1050 Brussels, Belgium
| | - Gianluca Bontempi
- Interuniversity Institute of Bioinformatics in Brussels (IB)2, 1050 Brussels, Belgium
- Machine Learning Group (MLG), Department d’Informatique, Universite libre de Bruxelles (ULB), 1050 Brussels, Belgium
| | - Isabella Castiglioni
- Institute of Molecular Bioimaging and Physiology, National Research Council (IBFM-CNR), Via F.Cervi 93, 20090 Milan, Segrate-Milan Italy
| |
Collapse
|
88
|
Szulzewsky F, Arora S, de Witte L, Ulas T, Markovic D, Schultze JL, Holland EC, Synowitz M, Wolf SA, Kettenmann H. Human glioblastoma-associated microglia/monocytes express a distinct RNA profile compared to human control and murine samples. Glia 2018; 64:1416-36. [PMID: 27312099 DOI: 10.1002/glia.23014] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Revised: 04/20/2016] [Accepted: 05/12/2016] [Indexed: 12/24/2022]
Abstract
Glioblastoma (GBM) is the most aggressive brain tumor in adults. It is strongly infiltrated by microglia and peripheral monocytes that support tumor growth. In the present study we used RNA sequencing to compare the expression profile of CD11b(+) human glioblastoma-associated microglia/monocytes (hGAMs) to CD11b(+) microglia isolated from non-tumor samples. Hierarchical clustering and principal component analysis showed a clear separation of the two sample groups and we identified 334 significantly regulated genes in hGAMs. In comparison to human control microglia hGAMs upregulated genes associated with mitotic cell cycle, cell migration, cell adhesion, and extracellular matrix organization. We validated the expression of several genes associated with extracellular matrix organization in samples of human control microglia, hGAMs, and the hGAMs-depleted fraction via qPCR. The comparison to murine GAMs (mGAMs) showed that both cell populations share a significant fraction of upregulated transcripts compared with their respective controls. These genes were mostly related to mitotic cell cycle. However, in contrast to murine cells, human GAMs did not upregulate genes associated to immune activation. Comparison of human and murine GAMs expression data to several data sets of in vitro-activated human macrophages and murine microglia showed that, in contrast to mGAMs, hGAMs share a smaller overlap to these data sets in general and in particular to cells activated by proinflammatory stimulation with LPS + INFγ or TNFα. Our findings provide new insights into the biology of human glioblastoma-associated microglia/monocytes and give detailed information about the validity of murine experimental models. GLIA 2016 GLIA 2016;64:1416-1436.
Collapse
Affiliation(s)
- Frank Szulzewsky
- Department of Cellular Neurosciences, Max-Delbrueck-Center for Molecular Medicine in the Helmholtz Society, Berlin, Germany.,Department of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Sonali Arora
- Department of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Lot de Witte
- Department of Psychiatry, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Thomas Ulas
- Department of Genomics and Immunoregulation, Life and Medical Sciences Institute University of Bonn, Bonn, Germany
| | - Darko Markovic
- Department of Cellular Neurosciences, Max-Delbrueck-Center for Molecular Medicine in the Helmholtz Society, Berlin, Germany.,Department of Neurosurgery, Helios Clinics, Berlin, Germany
| | - Joachim L Schultze
- Department of Genomics and Immunoregulation, Life and Medical Sciences Institute University of Bonn, Bonn, Germany.,Platform for Single Cell Genomics and Epigenomics at the German Center for Neurodegenerative Diseases and the University of Bonn, Bonn, Germany
| | - Eric C Holland
- Department of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Michael Synowitz
- Department of Cellular Neurosciences, Max-Delbrueck-Center for Molecular Medicine in the Helmholtz Society, Berlin, Germany.,Department of Neurosurgery, University Medical Center Schleswig-Holstein (UKSH), Kiel, Germany
| | - Susanne A Wolf
- Department of Cellular Neurosciences, Max-Delbrueck-Center for Molecular Medicine in the Helmholtz Society, Berlin, Germany
| | - Helmut Kettenmann
- Department of Cellular Neurosciences, Max-Delbrueck-Center for Molecular Medicine in the Helmholtz Society, Berlin, Germany
| |
Collapse
|
89
|
The Notch Interactome: Complexity in Signaling Circuitry. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1066:125-140. [PMID: 30030825 DOI: 10.1007/978-3-319-89512-3_7] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The Notch pathway controls a very broad spectrum of cell fates in metazoans during development, influencing proliferation, differentiation and cell death. Given its central role in normal development and homeostasis, misregulation of Notch signals can lead to various disorders including cancer. How the Notch pathway mediates such pleiotropic and differential effects is of fundamental importance. It is becoming increasingly clear through a number of large-scale genetic and proteomic studies that Notch interacts with a staggeringly large number of other genes and pathways in a context-dependent, complex, and highly regulated network, which determines the ultimate biological outcome. How best to interpret and analyze the continuously increasing wealth of data on Notch interactors remains a challenge. Here we review the current state of genetic and proteomic data related to the Notch interactome.
Collapse
|
90
|
Aung KL, Fischer SE, Denroche RE, Jang GH, Dodd A, Creighton S, Southwood B, Liang SB, Chadwick D, Zhang A, O'Kane GM, Albaba H, Moura S, Grant RC, Miller JK, Mbabaali F, Pasternack D, Lungu IM, Bartlett JMS, Ghai S, Lemire M, Holter S, Connor AA, Moffitt RA, Yeh JJ, Timms L, Krzyzanowski PM, Dhani N, Hedley D, Notta F, Wilson JM, Moore MJ, Gallinger S, Knox JJ. Genomics-Driven Precision Medicine for Advanced Pancreatic Cancer: Early Results from the COMPASS Trial. Clin Cancer Res 2017; 24:1344-1354. [PMID: 29288237 DOI: 10.1158/1078-0432.ccr-17-2994] [Citation(s) in RCA: 388] [Impact Index Per Article: 48.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Revised: 12/04/2017] [Accepted: 12/21/2017] [Indexed: 12/14/2022]
Abstract
Purpose: To perform real-time whole genome sequencing (WGS) and RNA sequencing (RNASeq) of advanced pancreatic ductal adenocarcinoma (PDAC) to identify predictive mutational and transcriptional features for better treatment selection.Experimental Design: Patients with advanced PDAC were prospectively recruited prior to first-line combination chemotherapy. Fresh tumor tissue was acquired by image-guided percutaneous core biopsy for WGS and RNASeq. Laser capture microdissection was performed for all cases. Primary endpoint was feasibility to report WGS results prior to first disease assessment CT scan at 8 weeks. The main secondary endpoint was discovery of patient subsets with predictive mutational and transcriptional signatures.Results: Sixty-three patients underwent a tumor biopsy between December 2015 and June 2017. WGS and RNASeq were successful in 62 (98%) and 60 (95%), respectively. Genomic results were reported at a median of 35 days (range, 19-52 days) from biopsy, meeting the primary feasibility endpoint. Objective responses to first-line chemotherapy were significantly better in patients with the classical PDAC RNA subtype compared with those with the basal-like subtype (P = 0.004). The best progression-free survival was observed in those with classical subtype treated with m-FOLFIRINOX. GATA6 expression in tumor measured by RNA in situ hybridization was found to be a robust surrogate biomarker for differentiating classical and basal-like PDAC subtypes. Potentially actionable genetic alterations were found in 30% of patients.Conclusions: Prospective genomic profiling of advanced PDAC is feasible, and our early data indicate that chemotherapy response differs among patients with different genomic/transcriptomic subtypes. Clin Cancer Res; 24(6); 1344-54. ©2017 AACR.
Collapse
Affiliation(s)
- Kyaw L Aung
- Wallace McCain Centre for Pancreatic Cancer, Department of Medical Oncology, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Sandra E Fischer
- Department of Pathology, University Health Network, University of Toronto, Toronto, Ontario, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Robert E Denroche
- PanCuRx Translational Research Initiative, Ontario, Institute for Cancer Research, Toronto, Ontario, Canada
| | - Gun-Ho Jang
- PanCuRx Translational Research Initiative, Ontario, Institute for Cancer Research, Toronto, Ontario, Canada
| | - Anna Dodd
- Wallace McCain Centre for Pancreatic Cancer, Department of Medical Oncology, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Sean Creighton
- Wallace McCain Centre for Pancreatic Cancer, Department of Medical Oncology, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Bernadette Southwood
- Wallace McCain Centre for Pancreatic Cancer, Department of Medical Oncology, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Sheng-Ben Liang
- UHN Biobank, University Health Network, Toronto, Ontario, Canada
| | - Dianne Chadwick
- UHN Biobank, University Health Network, Toronto, Ontario, Canada
| | - Amy Zhang
- PanCuRx Translational Research Initiative, Ontario, Institute for Cancer Research, Toronto, Ontario, Canada
| | - Grainne M O'Kane
- Wallace McCain Centre for Pancreatic Cancer, Department of Medical Oncology, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Hamzeh Albaba
- Wallace McCain Centre for Pancreatic Cancer, Department of Medical Oncology, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Shari Moura
- Wallace McCain Centre for Pancreatic Cancer, Department of Medical Oncology, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Robert C Grant
- Wallace McCain Centre for Pancreatic Cancer, Department of Medical Oncology, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Jessica K Miller
- Genomics, Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | - Faridah Mbabaali
- Genomics, Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | | | - Ilinca M Lungu
- Diagnostic Development, Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | - John M S Bartlett
- Diagnostic Development, Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | - Sangeet Ghai
- Joint Department of Medical Imaging, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Mathieu Lemire
- PanCuRx Translational Research Initiative, Ontario, Institute for Cancer Research, Toronto, Ontario, Canada
| | - Spring Holter
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Ashton A Connor
- PanCuRx Translational Research Initiative, Ontario, Institute for Cancer Research, Toronto, Ontario, Canada
| | - Richard A Moffitt
- Department of Pharmacology and Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina.,Department of Biomedical Informatics, Stony Brook University, Stony Brook, New York
| | - Jen Jen Yeh
- Department of Pharmacology and Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina.,Department of Surgery, University of North Carolina, Chapel Hill, North Carolina
| | - Lee Timms
- Genomics, Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | | | - Neesha Dhani
- Wallace McCain Centre for Pancreatic Cancer, Department of Medical Oncology, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - David Hedley
- Wallace McCain Centre for Pancreatic Cancer, Department of Medical Oncology, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Faiyaz Notta
- PanCuRx Translational Research Initiative, Ontario, Institute for Cancer Research, Toronto, Ontario, Canada
| | - Julie M Wilson
- PanCuRx Translational Research Initiative, Ontario, Institute for Cancer Research, Toronto, Ontario, Canada
| | - Malcolm J Moore
- British Columbia Cancer Agency, Vancouver, British Columbia, Canada
| | - Steven Gallinger
- Wallace McCain Centre for Pancreatic Cancer, Department of Medical Oncology, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, Ontario, Canada.,PanCuRx Translational Research Initiative, Ontario, Institute for Cancer Research, Toronto, Ontario, Canada.,Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada.,Hepatobiliary/Pancreatic Surgical Oncology Program, University Health Network, Toronto, Ontario, Canada
| | - Jennifer J Knox
- Wallace McCain Centre for Pancreatic Cancer, Department of Medical Oncology, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
91
|
Pietraszkiewicz A, van Asten F, Kwong A, Ratnapriya R, Abecasis G, Swaroop A, Chew EY. Association of Rare Predicted Loss-of-Function Variants in Cellular Pathways with Sub-Phenotypes in Age-Related Macular Degeneration. Ophthalmology 2017; 125:398-406. [PMID: 29224928 DOI: 10.1016/j.ophtha.2017.10.027] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Revised: 09/29/2017] [Accepted: 10/17/2017] [Indexed: 11/17/2022] Open
Abstract
PURPOSE To investigate the association of rare predicted loss-of-function (pLoF) variants within age-related macular degeneration (AMD) risk loci and AMD sub-phenotypes. DESIGN Case-control study. PARTICIPANTS Participants of AREDS, AREDS2, and Michigan Genomics Initiative. METHODS Whole genome sequencing data were analyzed for rare pLoF variants (frequency <0.1%) in the regions of previously identified 52 independent risk variants known to be associated with AMD. Frequency of the rare pLoF variants in cases with intermediate or advanced AMD was compared with controls. Variants were assigned to the complement, extracellular matrix (ECM), lipid, cell survival, immune system, metabolism, or unknown/other pathway. Associations of rare pLoF variant pathways with AMD sub-phenotypes were analyzed using logistic and linear regression, and Cox proportional hazards models. MAIN OUTCOME MEASURES Differences in rare pLoF variant pathway burden and association of rare pLoF variant pathways with sub-phenotypes within the population with AMD were evaluated. RESULTS Rare pLoF variants were found in 298 of 1689 cases (17.6%) and 237 of 1518 controls (15.6%) (odds ratio [OR], 1.11; 95% confidence interval [CI], 0.91-1.36; P = 0.310). An enrichment of rare pLoF variants in the complement pathway in cases versus controls (OR, 2.94; 95% CI, 1.49-5.79; P = 0.002) was observed. Within cases, associations between all rare pLoF variants and choroidal neovascularization (CNV) (OR, 1.34; 95% CI, 1.04-1.73; P = 0.023), calcified drusen (OR, 1.33; 95% CI, 1.04-1.72; P = 0.025), higher scores on the AREDS Extended AMD Severity Scale (Standardized Coefficient Beta (β)=0.346 [0.086-0.605], P = 0.009), and progression to advanced disease (hazard ratio, 1.25; 95% CI, 1.01-1.55; P = 0.042) were observed. At the pathway level, there were associations between the complement pathway and geographic atrophy (GA) (OR, 2.17; 95% CI, 1.12-4.24; P = 0.023), the complement pathway and calcified drusen (OR, 3.75; 95% CI, 1.79-7.86; P < 0.001), and the ECM pathway and more severe levels in the AREDS Extended AMD Severity Scale (β = 0.62; 95% CI, 0.04-1.20; P = 0.035). CONCLUSIONS Rare pLoF variants are associated with disease progression. Variants in the complement pathway modify the clinical course of AMD and increase the risk of developing specific sub-phenotypes.
Collapse
Affiliation(s)
- Alexandra Pietraszkiewicz
- Neurobiology, Neurodegeneration and Repair Laboratory, National Eye Institute, National Institutes of Health, Bethesda, Maryland
| | - Freekje van Asten
- Neurobiology, Neurodegeneration and Repair Laboratory, National Eye Institute, National Institutes of Health, Bethesda, Maryland; Division of Epidemiology and Clinical Applications, National Eye Institute, National Institutes of Health, Bethesda, Maryland
| | - Alan Kwong
- Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, Michigan
| | - Rinki Ratnapriya
- Neurobiology, Neurodegeneration and Repair Laboratory, National Eye Institute, National Institutes of Health, Bethesda, Maryland
| | - Goncalo Abecasis
- Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, Michigan
| | - Anand Swaroop
- Neurobiology, Neurodegeneration and Repair Laboratory, National Eye Institute, National Institutes of Health, Bethesda, Maryland
| | - Emily Y Chew
- Division of Epidemiology and Clinical Applications, National Eye Institute, National Institutes of Health, Bethesda, Maryland.
| |
Collapse
|
92
|
Bauer J, Grimm D, Gombocz E. Semantic analysis of thyroid cancer cell proteins obtained from rare research opportunities. J Biomed Inform 2017; 76:138-153. [DOI: 10.1016/j.jbi.2017.10.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Revised: 09/12/2017] [Accepted: 10/17/2017] [Indexed: 12/22/2022]
|
93
|
Tang S, Deng S, Guo J, Chen X, Zhang W, Cui Y, Luo Y, Yan Z, He QY, Shen S, Wang T. Deep Coverage Tissue and Cellular Proteomics Revealed IL-1β Can Independently Induce the Secretion of TNF-Associated Proteins from Human Synoviocytes. THE JOURNAL OF IMMUNOLOGY 2017; 200:821-833. [DOI: 10.4049/jimmunol.1700480] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Accepted: 10/31/2017] [Indexed: 01/15/2023]
|
94
|
Soh KP, Szczurek E, Sakoparnig T, Beerenwinkel N. Predicting cancer type from tumour DNA signatures. Genome Med 2017; 9:104. [PMID: 29183400 PMCID: PMC5706302 DOI: 10.1186/s13073-017-0493-2] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Accepted: 11/10/2017] [Indexed: 12/13/2022] Open
Abstract
Background Establishing the cancer type and site of origin is important in determining the most appropriate course of treatment for cancer patients. Patients with cancer of unknown primary, where the site of origin cannot be established from an examination of the metastatic cancer cells, typically have poor survival. Here, we evaluate the potential and limitations of utilising gene alteration data from tumour DNA to identify cancer types. Methods Using sequenced tumour DNA downloaded via the cBioPortal for Cancer Genomics, we collected the presence or absence of calls for gene alterations for 6640 tumour samples spanning 28 cancer types, as predictive features. We employed three machine-learning techniques, namely linear support vector machines with recursive feature selection, L1-regularised logistic regression and random forest, to select a small subset of gene alterations that are most informative for cancer-type prediction. We then evaluated the predictive performance of the models in a comparative manner. Results We found the linear support vector machine to be the most predictive model of cancer type from gene alterations. Using only 100 somatic point-mutated genes for prediction, we achieved an overall accuracy of 49.4±0.4 % (95 % confidence interval). We observed a marked increase in the accuracy when copy number alterations are included as predictors. With a combination of somatic point mutations and copy number alterations, a mere 50 genes are enough to yield an overall accuracy of 77.7±0.3 %. Conclusions A general cancer diagnostic tool that utilises either only somatic point mutations or only copy number alterations is not sufficient for distinguishing a broad range of cancer types. The combination of both gene alteration types can dramatically improve the performance. Electronic supplementary material The online version of this article (doi:10.1186/s13073-017-0493-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Kee Pang Soh
- Department of Biosystems Science and Engineering, ETH Zurich, Mattenstrasse 26, Basel, 4058, Switzerland.,Saw Swee Hock School of Public Health, National University of Singapore, Tahir Foundation Building, 12 Science Drive 2 MD1, Singapore, 117549, Singapore
| | - Ewa Szczurek
- Institute of Informatics, University of Warsaw, Banacha 2, Warsaw, 02-097, Poland
| | - Thomas Sakoparnig
- Biozentrum, University of Basel, Klingelbergstrasse 50/70, Basel, 4056, Switzerland.,SIB Swiss Institute of Bioinformatics, Basel, 4058, Switzerland
| | - Niko Beerenwinkel
- Department of Biosystems Science and Engineering, ETH Zurich, Mattenstrasse 26, Basel, 4058, Switzerland. .,SIB Swiss Institute of Bioinformatics, Basel, 4058, Switzerland.
| |
Collapse
|
95
|
Nyholt DR, Borsook D, Griffiths LR. Migrainomics — identifying brain and genetic markers of migraine. Nat Rev Neurol 2017; 13:725-741. [DOI: 10.1038/nrneurol.2017.151] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
96
|
Roncaglia P, van Dam TJP, Christie KR, Nacheva L, Toedt G, Huynen MA, Huntley RP, Gibson TJ, Lomax J. The Gene Ontology of eukaryotic cilia and flagella. Cilia 2017; 6:10. [PMID: 29177046 PMCID: PMC5688719 DOI: 10.1186/s13630-017-0054-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Accepted: 10/30/2017] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Recent research into ciliary structure and function provides important insights into inherited diseases termed ciliopathies and other cilia-related disorders. This wealth of knowledge needs to be translated into a computational representation to be fully exploitable by the research community. To this end, members of the Gene Ontology (GO) and SYSCILIA Consortia have worked together to improve representation of ciliary substructures and processes in GO. METHODS Members of the SYSCILIA and Gene Ontology Consortia suggested additions and changes to GO, to reflect new knowledge in the field. The project initially aimed to improve coverage of ciliary parts, and was then broadened to cilia-related biological processes. Discussions were documented in a public tracker. We engaged the broader cilia community via direct consultation and by referring to the literature. Ontology updates were implemented via ontology editing tools. RESULTS So far, we have created or modified 127 GO terms representing parts and processes related to eukaryotic cilia/flagella or prokaryotic flagella. A growing number of biological pathways are known to involve cilia, and we continue to incorporate this knowledge in GO. The resulting expansion in GO allows more precise representation of experimentally derived knowledge, and SYSCILIA and GO biocurators have created 199 annotations to 50 human ciliary proteins. The revised ontology was also used to curate mouse proteins in a collaborative project. The revised GO and annotations, used in comparative 'before and after' analyses of representative ciliary datasets, improve enrichment results significantly. CONCLUSIONS Our work has resulted in a broader and deeper coverage of ciliary composition and function. These improvements in ontology and protein annotation will benefit all users of GO enrichment analysis tools, as well as the ciliary research community, in areas ranging from microscopy image annotation to interpretation of high-throughput studies. We welcome feedback to further enhance the representation of cilia biology in GO.
Collapse
Affiliation(s)
- Paola Roncaglia
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SD UK
- The Gene Ontology Consortium, http://geneontology.org
| | - Teunis J. P. van Dam
- Centre for Molecular and Biomolecular Informatics, Radboud University Medical Center, PO Box 9101, 6500 HB Nijmegen, The Netherlands
- Theoretical Biology and Bioinformatics, Department of Biology, Faculty of Science, Utrecht University, Utrecht, The Netherlands
| | - Karen R. Christie
- The Gene Ontology Consortium, http://geneontology.org
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME 04609 USA
| | - Lora Nacheva
- Fakultät Biowissenschaften, Universität Heidelberg, Im Neuenheimer Feld 234, 69120 Heidelberg, Germany
| | - Grischa Toedt
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Meyerhofstr. 1, 69117 Heidelberg, Germany
| | - Martijn A. Huynen
- Centre for Molecular and Biomolecular Informatics, Radboud University Medical Center, PO Box 9101, 6500 HB Nijmegen, The Netherlands
| | - Rachael P. Huntley
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SD UK
- Present Address: Centre for Cardiovascular Genetics, University College London, London, WC1E 6JF UK
| | - Toby J. Gibson
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Meyerhofstr. 1, 69117 Heidelberg, Germany
| | - Jane Lomax
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SD UK
- The Gene Ontology Consortium, http://geneontology.org
- Present Address: SciBite Limited, BioData Innovation Centre, Wellcome Genome Campus, Cambridge, CB10 1DR UK
| |
Collapse
|
97
|
Combining membrane proteomics and computational three-way pathway analysis revealed signalling pathways preferentially regulated in human iPSCs and human ESCs. Sci Rep 2017; 7:15055. [PMID: 29118436 PMCID: PMC5678157 DOI: 10.1038/s41598-017-15347-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Accepted: 10/25/2017] [Indexed: 12/18/2022] Open
Abstract
Owing to the clinical potential of human induced pluripotent stem cells (hiPSCs) in regenerative medicine, a thorough examination of the similarities and differences between hiPSCs and human embryonic stem cells (hESCs) has become indispensable. Moreover, as the important roles of membrane proteins in biological signalling, functional analyses of membrane proteome are therefore promising. In this study, a pathway analysis by the bioinformatics tool GSEA was first performed to identify significant pathways associated with the three comparative membrane proteomics experiments: hiPSCs versus precursor human foreskin fibroblasts (HFF), hESCs versus precursor HFF, and hiPSCs versus hESCs. A following three-way pathway comparison was conducted to identify the differentially regulated pathways that may contribute to the differences between hiPSCs and hESCs. Our results revealed that pathways related to oxidative phosphorylation and focal adhesion may undergo incomplete regulations during the reprogramming process. This hypothesis was supported by another public proteomics dataset to a certain degree. The identified pathways and their core enriched proteins could serve as the starting point to explore the possible ways to make hiPSCs closer to hESCs.
Collapse
|
98
|
Transcriptomics-driven lipidomics (TDL) identifies the microbiome-regulated targets of ileal lipid metabolism. NPJ Syst Biol Appl 2017; 3:33. [PMID: 29138692 PMCID: PMC5676686 DOI: 10.1038/s41540-017-0033-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Revised: 08/28/2017] [Accepted: 09/13/2017] [Indexed: 12/31/2022] Open
Abstract
The gut microbiome and lipid metabolism are both recognized as essential components in the maintenance of metabolic health. The mechanisms involved are multifactorial and (especially for microbiome) poorly defined. A strategic approach to investigate the complexity of the microbial influence on lipid metabolism would facilitate determination of relevant molecular mechanisms for microbiome-targeted therapeutics. E. coli is associated with obesity and metabolic syndrome and we used this association in conjunction with gnotobiotic models to investigate the impact of E. coli on lipid metabolism. To address the complexities of the integration of the microbiome and lipid metabolism, we developed transcriptomics-driven lipidomics (TDL) to predict the impact of E. coli colonization on lipid metabolism and established mediators of inflammation and insulin resistance including arachidonic acid metabolism, alterations in bile acids and dietary lipid absorption. A microbiome-related therapeutic approach targeting these mechanisms may therefore provide a therapeutic avenue supporting maintenance of metabolic health. Microbes multifactorially impact host lipid metabolism bearing a significant impact in health and disease. A team led by Mojgan Masoodi and Scott Parkinson at Nestlé Institute of Health Sciences (NIHS) developed an integrative data driven approach for predictive lipidomics investigations of host-microbial impacts on lipid metabolism. Results of in-vivo studies with germ-free mice inoculated with E. coli and in-vitro studies demonstrated the multifactorial nature of the impact of E. coli on arachidonic acid metabolism in the ileum and altered host inflammation and lipid absorption. The findings provide insights into understanding the host-microbiome interactions and identifying microbiome-related solutions for maintaining health and tackling disease. The systems approach presented is applicable to investigate broad range of microbiome dependent and independent alterations in host lipid metabolism.
Collapse
|
99
|
Deeter A, Dalman M, Haddad J, Duan ZH. Inferring gene and protein interactions using PubMed citations and consensus Bayesian networks. PLoS One 2017; 12:e0186004. [PMID: 29049295 PMCID: PMC5648141 DOI: 10.1371/journal.pone.0186004] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Accepted: 09/22/2017] [Indexed: 11/25/2022] Open
Abstract
The PubMed database offers an extensive set of publication data that can be useful, yet inherently complex to use without automated computational techniques. Data repositories such as the Genomic Data Commons (GDC) and the Gene Expression Omnibus (GEO) offer experimental data storage and retrieval as well as curated gene expression profiles. Genetic interaction databases, including Reactome and Ingenuity Pathway Analysis, offer pathway and experiment data analysis using data curated from these publications and data repositories. We have created a method to generate and analyze consensus networks, inferring potential gene interactions, using large numbers of Bayesian networks generated by data mining publications in the PubMed database. Through the concept of network resolution, these consensus networks can be tailored to represent possible genetic interactions. We designed a set of experiments to confirm that our method is stable across variation in both sample and topological input sizes. Using gene product interactions from the KEGG pathway database and data mining PubMed publication abstracts, we verify that regardless of the network resolution or the inferred consensus network, our method is capable of inferring meaningful gene interactions through consensus Bayesian network generation with multiple, randomized topological orderings. Our method can not only confirm the existence of currently accepted interactions, but has the potential to hypothesize new ones as well. We show our method confirms the existence of known gene interactions such as JAK-STAT-PI3K-AKT-mTOR, infers novel gene interactions such as RAS- Bcl-2 and RAS-AKT, and found significant pathway-pathway interactions between the JAK-STAT signaling and Cardiac Muscle Contraction KEGG pathways.
Collapse
Affiliation(s)
- Anthony Deeter
- Integrated Bioscience, University of Akron, Akron, Ohio, United States of America
- Department of Computer Science, University of Akron, Akron, Ohio, United States of America
- * E-mail:
| | - Mark Dalman
- College of Public Health, Department of Biostatistics, Environmental Health Sciences and Epidemiology, Kent State University, Kent, Ohio, United States of America
- College of Podiatric Medicine, Department of Preclinical Sciences, Kent State University, Kent, Ohio, United States of America
| | - Joseph Haddad
- Department of Computer Science, University of Akron, Akron, Ohio, United States of America
| | - Zhong-Hui Duan
- Integrated Bioscience, University of Akron, Akron, Ohio, United States of America
- Department of Computer Science, University of Akron, Akron, Ohio, United States of America
| |
Collapse
|
100
|
Ellison EM, Bradley-Whitman MA, Lovell MA. Single-Base Resolution Mapping of 5-Hydroxymethylcytosine Modifications in Hippocampus of Alzheimer's Disease Subjects. J Mol Neurosci 2017; 63:185-197. [PMID: 28866733 PMCID: PMC5909840 DOI: 10.1007/s12031-017-0969-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Accepted: 08/23/2017] [Indexed: 12/19/2022]
Abstract
Epigenetic modifications to cytosine have been shown to regulate transcription in cancer, embryonic development, and recently neurodegeneration. While cytosine methylation studies are now common in neurodegenerative research, hydroxymethylation studies are rare, particularly genome-wide mapping studies. As an initial study to analyze 5-hydroxymethylcytosine (5-hmC) in the Alzheimer's disease (AD) genome, reduced representation hydroxymethylation profiling (RRHP) was used to analyze more than 2 million sites of possible modification in hippocampal DNA of sporadic AD and normal control subjects. Genes with differentially hydroxymethylated regions were filtered based on previously published microarray data for altered gene expression in hippocampal DNA of AD subjects. Our data show significant pathways for altered levels of 5-hmC in the hippocampus of AD subjects compared to age-matched normal controls involved in signaling, energy metabolism, cell function, gene expression, protein degradation, and cell structure and stabilization. Overall, our data suggest a possible role for the dysregulation of epigenetic modifications to cytosine in late stage AD.
Collapse
Affiliation(s)
| | - Melissa A Bradley-Whitman
- Sanders-Brown Center on Aging, University of Kentucky, 135 Sanders-Brown Center on Aging, 800 South Limestone S, Lexington, KY, 40536, USA
| | - Mark A Lovell
- Department of Chemistry, University of Kentucky, Lexington, KY, USA.
- Sanders-Brown Center on Aging, University of Kentucky, 135 Sanders-Brown Center on Aging, 800 South Limestone S, Lexington, KY, 40536, USA.
| |
Collapse
|