12 results on '"Tristan M. Carland"'
Search Results
2. Group-based variant calling leveraging next-generation supercomputing for large-scale whole-genome sequencing studies.
- Author
-
Kristopher Standish, Tristan M. Carland, Glenn K. Lockwood, Wayne Pfeiffer, Mahidhar Tatineni, C. Huang, Sarah Lamberth, Yauheniya Cherkas, Carrie Brodmerkel, Ed Jaeger, Lance Smith, Gunaretnam Rajagopal, Mark Curran, and Nicholas J. Schork
- Published
- 2015
- Full Text
- View/download PDF
3. Marine Natural Product Honaucin A Attenuates Inflammation by Activating the Nrf2-ARE Pathway
- Author
-
Amro Hamdoun, Victor Nizet, Terry Gaasterland, Tristan M. Carland, William H. Gerwick, Joseph P. Campanale, Lena Gerwick, Samantha J. Mascuch, N. Tessa Pierce, Mary E. Hensler, Joshua Olson, Paul D. Boudreau, and Hyukjae Choi
- Subjects
0301 basic medicine ,Aquatic Organisms ,Alkylation ,Response element ,Anti-Inflammatory Agents ,Pharmaceutical Science ,Medical and Health Sciences ,Antioxidants ,Analytical Chemistry ,Mice ,Drug Discovery ,Gene expression ,Tumor ,Kelch-Like ECH-Associated Protein 1 ,Chemistry ,Biological Sciences ,Cell biology ,MCF-7 Cells ,Molecular Medicine ,Female ,Signal transduction ,medicine.symptom ,Signal Transduction ,Biotechnology ,NF-E2-Related Factor 2 ,1.1 Normal biological development and functioning ,Medicinal & Biomolecular Chemistry ,Repressor ,Article ,Cell Line ,03 medical and health sciences ,Underpinning research ,Cell Line, Tumor ,Complementary and Integrative Health ,medicine ,Genetics ,Animals ,Humans ,Nutrition ,Pharmacology ,Inflammation ,Biological Products ,Organic Chemistry ,KEAP1 ,In vitro ,Cytosol ,030104 developmental biology ,RAW 264.7 Cells ,Complementary and alternative medicine ,Mechanism of action ,Cytoprotection ,Chemical Sciences - Abstract
The cyanobacterial marine natural product honaucin A inhibits mammalian innate inflammation in vitro and in vivo. To decipher its mechanism of action, RNA sequencing was used to evaluate differences in gene expression of cultured macrophages following honaucin A treatment. This analysis led to the hypothesis that honaucin A exerts its anti-inflammatory activity through activation of the cytoprotective nuclear erythroid 2-related factor 2 (Nrf2)-antioxidant response element/electrophile response element (ARE/EpRE) signaling pathway. Activation of this pathway by honaucin A in cultured human MCF7 cells was confirmed using an Nrf2 luciferase reporter assay. In vitro alkylation experiments with the natural product and N-acetyl-L-cysteine suggest that honaucin A activates this pathway through covalent interaction with the sulfhydryl residues of the cytosolic repressor protein Keapl. Honaucin A presents a potential therapeutic lead for diseases with an inflammatory component modulated by Nrf2-ARE.
- Published
- 2018
4. p53-Dependent DNA damage response sensitive to editing-defective tRNA synthetase in zebrafish
- Author
-
Yi Shi, Steven R. Head, Tristan M. Carland, Shuji Kishi, Tomoyuki Sasaki, Shanshan Lian, Youngzee Song, Nicholas J. Schork, and Paul Schimmel
- Subjects
0301 basic medicine ,Male ,Embryo, Nonmammalian ,DNA repair ,DNA damage ,Biology ,medicine.disease_cause ,Amino Acyl-tRNA Synthetases ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,medicine ,Animals ,Zebrafish ,Genetics ,Mutation ,Multidisciplinary ,Gadd45 ,Gene Expression Regulation, Developmental ,Biological Sciences ,Zebrafish Proteins ,biology.organism_classification ,030104 developmental biology ,chemistry ,RNA editing ,030220 oncology & carcinogenesis ,Transfer RNA ,Female ,RNA Editing ,Tumor Suppressor Protein p53 ,DNA ,DNA Damage - Abstract
Brain and heart pathologies are caused by editing defects of transfer RNA (tRNA) synthetases, which preserve genetic code fidelity by removing incorrect amino acids misattached to tRNAs. To extend understanding of the broader impact of synthetase editing reactions on organismal homeostasis, and based on effects in bacteria ostensibly from small amounts of mistranslation of components of the replication apparatus, we investigated the sensitivity to editing of the vertebrate genome. We show here that in zebrafish embryos, transient overexpression of editing-defective valyl-tRNA synthetase (ValRS(ED)) activated DNA break-responsive H2AX and p53-responsive downstream proteins, such as cyclin-dependent kinase (CDK) inhibitor p21, which promotes cell-cycle arrest at DNA damage checkpoints, and Gadd45 and p53R2, with pivotal roles in DNA repair. In contrast, the response of these proteins to expression of ValRS(ED) was abolished in p53-deficient fish. The p53-activated downstream signaling events correlated with suppression of abnormal morphological changes caused by the editing defect and, in adults, reversed a shortened life span (followed for 2 y). Conversely, with normal editing activities, p53-deficient fish have a normal life span and few morphological changes. Whole-fish deep sequencing showed genomic mutations associated with the editing defect. We suggest that the sensitivity of p53 to expression of an editing-defective tRNA synthetase has a critical role in promoting genome integrity and organismal homeostasis.
- Published
- 2016
5. Differential expression and intrachromosomal evolution of the sghC1q genes in zebrafish (Danio rerio)
- Author
-
Tristan M. Carland, Jeffrey B. Locke, Victor Nizet, and Lena Gerwick
- Subjects
Immunology ,Danio ,Biology ,Genome ,Evolution, Molecular ,Transcription (biology) ,Phylogenetics ,Streptococcal Infections ,Chromosome Duplication ,Gene duplication ,Animals ,Zebrafish ,Gene ,Phylogeny ,Genetics ,Complement C1q ,Gene Expression Profiling ,Gene Expression Regulation, Developmental ,Streptococcus ,Zebrafish Proteins ,biology.organism_classification ,Immunity, Innate ,Up-Regulation ,Gene expression profiling ,Multigene Family ,Developmental Biology - Abstract
The secreted globular head C1q (sghC1q) genes can be characterized as a family of genetic loci encoding signal peptides followed by single complement component 1q globular (gC1q) motifs. Members of this family have been referred to as precerebellin-like (Cblnl), C1q-like or ovary specific C1q-like factors, and are transcribed in response to infection and/or during early development. This study was primarily undertaken to identify the zebrafish sghC1q (or DrsghC1q) genes that increase their transcription in response to infection and to examine their transcriptional patterns during early development. Twenty sghC1q genes were found in the zebrafish (Danio rerio) genome (Zv9). Two of the examined twenty genes showed significant up-regulation within 24 h of infection with the fish pathogen Streptococcus iniae, and eleven of the examined twenty were expressed during early development. Due to the clustered nature of these genes on chromosomes two and seven, intrachromosomal duplication events are hypothesized and explored.
- Published
- 2012
- Full Text
- View/download PDF
6. Honaucin A, mechanism of action and role as a potential cancer prevention agent
- Author
-
Paul D. Boudreau, Gabriel Navarro, SJ Mascuch, Terry Gaasterland, Lena Gerwick, Tristan M. Carland, and William H. Gerwick
- Subjects
Pharmacology ,Cancer prevention ,business.industry ,Organic Chemistry ,Pharmaceutical Science ,Bioinformatics ,Analytical Chemistry ,Complementary and alternative medicine ,Mechanism of action ,Drug Discovery ,medicine ,Molecular Medicine ,medicine.symptom ,business - Published
- 2015
- Full Text
- View/download PDF
7. Group-based variant calling leveraging next-generation supercomputing for large-scale whole-genome sequencing studies
- Author
-
Conway C. Huang, Glenn K. Lockwood, Nicholas J. Schork, S. Lamberth, Carrie Brodmerkel, Yauheniya Cherkas, Tristan M. Carland, Wayne Pfeiffer, Kristopher A. Standish, Gunaretnam Rajagopal, Ed Jaeger, Mark Curran, Lance Smith, and Mahidhar Tatineni
- Subjects
Big data ,Genomics ,Variation (game tree) ,Computational biology ,Biology ,Biochemistry ,Polymorphism, Single Nucleotide ,03 medical and health sciences ,0302 clinical medicine ,Structural Biology ,Variant calling ,Humans ,Molecular Biology ,030304 developmental biology ,030203 arthritis & rheumatology ,Whole genome sequencing ,0303 health sciences ,Whole-genome sequencing ,business.industry ,Computers ,Genome, Human ,Applied Mathematics ,Scale (chemistry) ,Methodology Article ,High-Throughput Nucleotide Sequencing ,Supercomputing ,Sequence Analysis, DNA ,Supercomputer ,Data science ,Computer Science Applications ,Workflow ,Data Interpretation, Statistical ,Human genome ,business ,Software - Abstract
Motivation Next-generation sequencing (NGS) technologies have become much more efficient, allowing whole human genomes to be sequenced faster and cheaper than ever before. However, processing the raw sequence reads associated with NGS technologies requires care and sophistication in order to draw compelling inferences about phenotypic consequences of variation in human genomes. It has been shown that different approaches to variant calling from NGS data can lead to different conclusions. Ensuring appropriate accuracy and quality in variant calling can come at a computational cost. Results We describe our experience implementing and evaluating a group-based approach to calling variants on large numbers of whole human genomes. We explore the influence of many factors that may impact the accuracy and efficiency of group-based variant calling, including group size, the biogeographical backgrounds of the individuals who have been sequenced, and the computing environment used. We make efficient use of the Gordon supercomputer cluster at the San Diego Supercomputer Center by incorporating job-packing and parallelization considerations into our workflow while calling variants on 437 whole human genomes generated as part of large association study. Conclusions We ultimately find that our workflow resulted in high-quality variant calls in a computationally efficient manner. We argue that studies like ours should motivate further investigations combining hardware-oriented advances in computing systems with algorithmic developments to tackle emerging ‘big data’ problems in biomedical research brought on by the expansion of NGS technologies. Electronic supplementary material The online version of this article (doi:10.1186/s12859-015-0736-4) contains supplementary material, which is available to authorized users.
- Published
- 2014
8. Omics Pipe: a community-based framework for reproducible multi-omics data analysis
- Author
-
Louis Gioia, Kathleen M. Fisch, Andrew I. Su, Salvatore Loguercio, Tristan M. Carland, Jean-Christophe Ducom, and Tobias Meißner
- Subjects
Statistics and Probability ,Databases, Factual ,Computer science ,Sequence analysis ,Genomics ,Breast Neoplasms ,computer.software_genre ,Biochemistry ,DNA sequencing ,RefSeq ,Cluster Analysis ,Humans ,Exome ,Molecular Biology ,Whole genome sequencing ,Sequence Analysis, RNA ,RNA ,High-Throughput Nucleotide Sequencing ,Reproducibility of Results ,Omics ,Original Papers ,Computer Science Applications ,Computational Mathematics ,Computational Theory and Mathematics ,Multi omics ,Female ,Data mining ,computer ,Software - Abstract
Motivation: Omics Pipe ( http://sulab.scripps.edu/omicspipe ) is a computational framework that automates multi-omics data analysis pipelines on high performance compute clusters and in the cloud. It supports best practice published pipelines for RNA-seq, miRNA-seq, Exome-seq, Whole-Genome sequencing, ChIP-seq analyses and automatic processing of data from The Cancer Genome Atlas (TCGA). Omics Pipe provides researchers with a tool for reproducible, open source and extensible next generation sequencing analysis. The goal of Omics Pipe is to democratize next-generation sequencing analysis by dramatically increasing the accessibility and reproducibility of best practice computational pipelines, which will enable researchers to generate biologically meaningful and interpretable results. Results: Using Omics Pipe, we analyzed 100 TCGA breast invasive carcinoma paired tumor-normal datasets based on the latest UCSC hg19 RefSeq annotation. Omics Pipe automatically downloaded and processed the desired TCGA samples on a high throughput compute cluster to produce a results report for each sample. We aggregated the individual sample results and compared them to the analysis in the original publications. This comparison revealed high overlap between the analyses, as well as novel findings due to the use of updated annotations and methods. Availability and implementation: Source code for Omics Pipe is freely available on the web ( https://bitbucket.org/sulab/omics_pipe ). Omics Pipe is distributed as a standalone Python package for installation ( https://pypi.python.org/pypi/omics_pipe ) and as an Amazon Machine Image in Amazon Web Services Elastic Compute Cloud that contains all necessary third-party software dependencies and databases ( https://pythonhosted.org/omics_pipe/AWS_installation.html ). Contact: asu@scripps.edu or kfisch@ucsd.edu Supplementary Information: Supplementary data are available at Bioinformatics online.
- Published
- 2014
9. Omics Pipe: A Computational Framework for Reproducible Multi-Omics Data Analysis
- Author
-
Kathleen M. Fisch, Andrew I. Su, Salvatore Loguercio, Tristan M. Carland, Tobias Meißner, Jean-Christophe Ducom, and Louis Gioia
- Subjects
Whole genome sequencing ,0303 health sciences ,Computer science ,Omics ,computer.software_genre ,DNA sequencing ,03 medical and health sciences ,0302 clinical medicine ,Cancer genome ,Multi omics ,Data mining ,computer ,030217 neurology & neurosurgery ,030304 developmental biology - Abstract
Omics Pipe (https://bitbucket.org/sulab/omics_pipe) is a computational platform that automates multi-omics data analysis pipelines on high performance compute clusters and in the cloud. It supports best practice published pipelines for RNA-seq, miRNA-seq, Exome-seq, Whole Genome sequencing, ChIP-seq analyses and automatic processing of data from The Cancer Genome Atlas. Omics Pipe provides researchers with a tool for reproducible, open source and extensible next generation sequencing analysis.
- Published
- 2014
- Full Text
- View/download PDF
10. The C1q domain containing proteins: Where do they come from and what do they do?
- Author
-
Tristan M. Carland and Lena Gerwick
- Subjects
Signal peptide ,Genetics ,Bodily Secretions ,Protein Folding ,EGF-like domain ,Complement C1q ,Immunology ,Immunity ,Computational Biology ,Biology ,Genome ,Protein Structure, Tertiary ,Evolution, Molecular ,Protein structure ,Phylogenetics ,C1q domain ,Animals ,Humans ,Protein folding ,Gene ,Phylogeny ,Developmental Biology - Abstract
The gene sequence encoding an N-terminal collagen stalk followed by a globular complement 1q domain (gC1q), an architecture that characterizes the C1q A, B and C chains of the first complement component (C1), did not become prevalent until the cephalochordates and urochordates. However, genes encoding only the globular complement 1q domain (ghC1q) are more ancient as they exist within many lower vertebrate and invertebrate genomes, and are even present in the prokaryotes. These genes can be divided into two groups, the first, which appears to be the more ancient form, encodes proteins that are not secreted (cghC1q). The second group encodes proteins in which the globular domain is preceded by a signal peptide indicating secretion (sghC1q). In this review we examine bioinformatic evidence for C1q domain containing (C1qDC) genes in many organisms and integrate these observations with research performed and published on the biochemistry and functions of this fascinating set of proteins.
- Published
- 2010
11. Transcriptional studies of a novel family of 'short C1q' domain proteins in zebrafish
- Author
-
Tristan M. Carland, Jeffrey B. Locke, Lena Gerwick, and Victor Nizet
- Subjects
biology ,Genetics ,C1q domain ,Computational biology ,biology.organism_classification ,Molecular Biology ,Biochemistry ,Zebrafish ,Molecular biology ,Biotechnology - Published
- 2008
- Full Text
- View/download PDF
12. Whole Genome Sequence Analysis Of A Cousin Pair With Restricting Anorexia Nervosa
- Author
-
Andrew W. Bergen, Walter H. Kaye, Nicholas J. Schork, Manfred M. Fichter, Vikas Bansal, Tristan M. Carland, Ashley A. Scott-Van Zeeland, Christine Strobel, Pierre J. Magistretti, and Pei-an Betty Shih
- Subjects
Pharmacology ,Whole genome sequencing ,Genetics ,Haplotype ,Genomics ,Genome-wide association study ,Single-nucleotide polymorphism ,Biology ,Phenotype ,Psychiatry and Mental health ,Neurology ,Anorexia nervosa (differential diagnoses) ,Pharmacology (medical) ,Neurology (clinical) ,Gene ,Biological Psychiatry - Abstract
Background Anorexia Nervosa (AN) is a serious mental illness that often has an onset during adolescence. AN is characterized by emaciation, fear of gaining weight despite being underweight, and the highest mortality rate of all psychiatric illnesses. AN is highly heritable and recent meta-analyses in 3,495 anorexia nervosa cases and 10,982 controls has identified a region on chromosome 12 at genome-wide significance, significant common genetic heritability, and genetic correlations with both psychiatric and metabolic traits. AN symptoms and personality traits tend to be present in unaffected family members of the patients, suggesting that certain shared genetic factors within each family may contribute to the unique risk underlying an affected individual's phenotype. Methods To gain insight into the role that unique, family-specific variants may play in the development of AN, we performed whole genome sequencing analysis (Complete Genomics) and extensive annotation of high quality variants (Cypher Genomics, SG-Advisor, ANNOVAR) to search for genetic variants that may influence AN risk in six individuals, consisting of two maternally-linked cousins with severe AN and their parents. We focused on maternally-inherited shared segments; the preferred transmission model for variant filtering was the dominant model. We reviewed results from the Psychiatric Genomic Consortium AN GWAS and other -omic databases at identified genes. Results Of the approximately 5.3 million variants per individual that were analyzed, 494,712 were shared Identical-By-Descent (IBD) by the cousin pair based on maternally derived haplotypes. Based on extensive genetic variant annotations within maternally inherited shared segments, we identified variants in TTC22, MRPS9, DNAJC30, HEPACAM2, USP20, ESF1, and CDK5RAP1. Multiple prediction methods suggest that six of these variants are likely to affect protein function. The region proximal to MRPS9 exhibits an excess of nominally significant findings in the PGC AN GWAS, and eQTLs to MRPS9 and to an uncharacterized antisense gene in many tissues. MRPS9 SNPs are associated in blood with 4-androsten-3beta,17beta-diol disulfate 2 and dehydroisoandrosterone sulfate (a metabolite and a precursor of steroid sex hormones, respectively). Discussion By capitalizing on the homogeneity of the disease presentation and the genomic makeup among two cousins with a diagnosis of restricting-type AN, we exploited whole genome sequence analysis and variant annotation approaches to identify a set of novel variants and genes that may influence AN. Our results suggest that there may be utility in whole genome sequencing of families with affected individuals to detect variants that contribute to a complex disease such as AN.
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.