Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Samir N. Kelada is active.

Publication


Featured researches published by Samir N. Kelada.


Genome Research | 2011

Genetic analysis of complex traits in the emerging Collaborative Cross

David L. Aylor; William Valdar; Wendy Foulds-Mathes; Ryan J. Buus; Ricardo A. Verdugo; Ralph S. Baric; Martin T. Ferris; Jeffrey A. Frelinger; Mark T. Heise; Matt Frieman; Lisa E. Gralinski; Timothy A. Bell; John D. Didion; Kunjie Hua; Derrick L. Nehrenberg; Christine L. Powell; Jill Steigerwalt; Yuying Xie; Samir N. Kelada; Francis S. Collins; Ivana V. Yang; David A. Schwartz; Lisa A. Branstetter; Elissa J. Chesler; Darla R. Miller; Jason S. Spence; Eric Yi Liu; Leonard McMillan; Abhishek Sarkar; Jeremy Wang

The Collaborative Cross (CC) is a mouse recombinant inbred strain panel that is being developed as a resource for mammalian systems genetics. Here we describe an experiment that uses partially inbred CC lines to evaluate the genetic properties and utility of this emerging resource. Genome-wide analysis of the incipient strains reveals high genetic diversity, balanced allele frequencies, and dense, evenly distributed recombination sites-all ideal qualities for a systems genetics resource. We map discrete, complex, and biomolecular traits and contrast two quantitative trait locus (QTL) mapping approaches. Analysis based on inferred haplotypes improves power, reduces false discovery, and provides information to identify and prioritize candidate genes that is unique to multifounder crosses like the CC. The number of expression QTLs discovered here exceeds all previous efforts at eQTL mapping in mice, and we map local eQTL at 1-Mb resolution. We demonstrate that the genetic diversity of the CC, which derives from random mixing of eight founder strains, results in high phenotypic diversity and enhances our ability to map causative loci underlying complex disease-related traits.


Neuroscience Letters | 2003

Glutathione S-transferase M1, T1, and P1 polymorphisms and Parkinson's disease.

Samir N. Kelada; Patricia L. Stapleton; Federico M. Farin; Theo K. Bammler; David L. Eaton; Terri Smith-Weller; Gary M. Franklin; Phillip D. Swanson; William T. Longstreth; Harvey Checkoway

Oxidative stress is widely thought to contribute significantly to the pathogenesis Parkinsons disease (PD). Given the role of glutathione S-transferases (GSTs) in the conjugation of electrophiles and protection against reactive oxygen species, genes encoding the GSTs have been considered candidates for association studies of PD. We tested for associations between genotypes of GSTM1(homozygous deletion vs. non-deleted), GSTT1(homozygous deletion vs. non-deleted), and GSTP1 (Ile104Val and Ala113Val) and PD in a case-control study of 214 idiopathic PD cases and 330 age- and gender-matched, unrelated controls of Caucasian ethnicity. No significant associations with any of the GST genotypes were observed. However, there was a marginally significant difference in the distribution of GSTP1 104 genotypes between cases and controls (P=0.07), with an excess of Ile104Val heterozygotes found among cases (odds ratio (OR)=1.43; 95% Confidence Interval (CI): 0.98-2.08). This difference in the genotype distribution was strongest among smokers (OR for heterozygote=1.92; 95% CI: 1.12-3.29) versus non-smokers and among males (OR for heterozygote=1.99; 95% CI: 1.24-3.19) versus females. The distribution of GSTP1 Ile104Val and Ala113Val haplotypes did not differ between cases and controls. Taken together, these results suggest a potentially minor role of GSTP1 in PD, but do not give evidence for associations with either GSTM1 or GSTT1.


PLOS Pathogens | 2013

miR-182 and miR-10a are key regulators of Treg specialisation and stability during Schistosome and Leishmania-associated inflammation.

Samir N. Kelada; Praveen Sethupathy; Isobel S. Okoye; Eleni Kistasis; Stephanie Czieso; Sandra White; David Chou; Craig Martens; Stacy Ricklefs; Kimmo Virtaneva; Dan Sturdevant; Stephen F. Porcella; Yasmine Belkaid; Thomas A. Wynn; Mark S. Wilson

A diverse suite of effector immune responses provide protection against various pathogens. However, the array of effector responses must be immunologically regulated to limit pathogen- and immune-associated damage. CD4+Foxp3+ regulatory T cells (Treg) calibrate immune responses; however, how Treg cells adapt to control different effector responses is unclear. To investigate the molecular mechanism of Treg diversity we used whole genome expression profiling and next generation small RNA sequencing of Treg cells isolated from type-1 or type-2 inflamed tissue following Leishmania major or Schistosoma mansoni infection, respectively. In-silico analyses identified two miRNA “regulatory hubs” miR-10a and miR-182 as critical miRNAs in Th1- or Th2-associated Treg cells, respectively. Functionally and mechanistically, in-vitro and in-vivo systems identified that an IL-12/IFNγ axis regulated miR-10a and its putative transcription factor, Creb. Importantly, reduced miR-10a in Th1-associated Treg cells was critical for Treg function and controlled a suite of genes preventing IFNγ production. In contrast, IL-4 regulated miR-182 and cMaf in Th2-associed Treg cells, which mitigated IL-2 secretion, in part through repression of IL2-promoting genes. Together, this study indicates that CD4+Foxp3+ cells can be shaped by local environmental factors, which orchestrate distinct miRNA pathways preserving Treg stability and suppressor function.


G3: Genes, Genomes, Genetics | 2012

Genetic Analysis of Hematological Parameters in Incipient Lines of the Collaborative Cross

Samir N. Kelada; David L. Aylor; Bailey C. Peck; Joseph F. Ryan; Urraca Tavarez; Ryan J. Buus; Darla R. Miller; Elissa J. Chesler; David W. Threadgill; Gary A. Churchill; Fernando Pardo-Manuel de Villena; Francis S. Collins

Hematological parameters, including red and white blood cell counts and hemoglobin concentration, are widely used clinical indicators of health and disease. These traits are tightly regulated in healthy individuals and are under genetic control. Mutations in key genes that affect hematological parameters have important phenotypic consequences, including multiple variants that affect susceptibility to malarial disease. However, most variation in hematological traits is continuous and is presumably influenced by multiple loci and variants with small phenotypic effects. We used a newly developed mouse resource population, the Collaborative Cross (CC), to identify genetic determinants of hematological parameters. We surveyed the eight founder strains of the CC and performed a mapping study using 131 incipient lines of the CC. Genome scans identified quantitative trait loci for several hematological parameters, including mean red cell volume (Chr 7 and Chr 14), white blood cell count (Chr 18), percent neutrophils/lymphocytes (Chr 11), and monocyte number (Chr 1). We used evolutionary principles and unique bioinformatics resources to reduce the size of candidate intervals and to view functional variation in the context of phylogeny. Many quantitative trait loci regions could be narrowed sufficiently to identify a small number of promising candidate genes. This approach not only expands our knowledge about hematological traits but also demonstrates the unique ability of the CC to elucidate the genetic architecture of complex traits.


American Journal of Respiratory Cell and Molecular Biology | 2011

Strain-dependent genomic factors affect allergen-induced airway hyperresponsiveness in mice.

Samir N. Kelada; Mark S. Wilson; Urraca Tavarez; Kari Kubalanza; Bhavesh Borate; Greg S. Whitehead; Shuichiro Maruoka; Michelle G. Roy; Michelle Olive; Danielle E. Carpenter; David M. Brass; Thomas A. Wynn; Donald N. Cook; Christopher M. Evans; David A. Schwartz; Francis S. Collins

Asthma is etiologically and clinically heterogeneous, making the genomic basis of asthma difficult to identify. We exploited the strain-dependence of a murine model of allergic airway disease to identify different genomic responses in the lung. BALB/cJ and C57BL/6J mice were sensitized with the immunodominant allergen from the Dermatophagoides pteronyssinus species of house dust mite (Der p 1), without exogenous adjuvant, and the mice then underwent a single challenge with Der p 1. Allergic inflammation, serum antibody titers, mucous metaplasia, and airway hyperresponsiveness were evaluated 72 hours after airway challenge. Whole-lung gene expression analyses were conducted to identify genomic responses to allergen challenge. Der p 1-challenged BALB/cJ mice produced all the key features of allergic airway disease. In comparison, C57BL/6J mice produced exaggerated Th2-biased responses and inflammation, but exhibited an unexpected decrease in airway hyperresponsiveness compared with control mice. Lung gene expression analysis revealed genes that were shared by both strains and a set of down-regulated genes unique to C57BL/6J mice, including several G-protein-coupled receptors involved in airway smooth muscle contraction, most notably the M2 muscarinic receptor, which we show is expressed in airway smooth muscle and was decreased at the protein level after challenge with Der p 1. Murine strain-dependent genomic responses in the lung offer insights into the different biological pathways that develop after allergen challenge. This study of two different murine strains demonstrates that inflammation and airway hyperresponsiveness can be decoupled, and suggests that the down-modulation of expression of G-protein-coupled receptors involved in regulating airway smooth muscle contraction may contribute to this dissociation.


Neurotoxicology | 2002

Gender difference in the interaction of smoking and monoamine oxidase B intron 13 genotype in Parkinson's disease.

Samir N. Kelada; Paola Costa-Mallen; Lucio G. Costa; Terry Smith-Weller; Gary M. Franklin; Phillip D. Swanson; William T. Longstreth; Harvey Checkoway

We tested for gender-specific interactions between smoking and genetic polymorphisms of monoamine oxidase B (MAO-B) intron 13 (G or A allele), monoamine oxidase A (MAO-A) EcoRV (Yor N allele), and dopamine D2 recepor (DRD2) Taq1B (B1 or B2 allele) in a case-control study of 186 incident idiopathic Parkinsons disease (PD) cases and 296 age- and gender-matched controls. The odds ratios (ORs) for PD risk for ever smokers versus never smokers were 0.27 (95% CI: 0.13-0.58) for men of genotype G, and 1.26 (0.60-2.63) for men of genotype A (interaction chi2 = 8.14, P = 0.004). In contrast, for women, the OR for ever smokers versus never smokers were 0.62 (95% CI: 0.25-1.34) and 0.64 (95% CI: 0.18-2.21) for women of genotype GG/GA and AA, respectively (interaction chi2 = 0.001, P = 0.975). No interactions were detected between smoking and either MAO-A EcoRV or DRD2 Taq1B genotypes. These results suggest that a strong gender difference exists with respect to the modifying effect of MAO-B genotype on the smoking association with PD.


Translational Psychiatry | 2014

Transcriptional targets of the schizophrenia risk gene MIR137

Ann L. Collins; Y Kim; Rachael J. Bloom; Samir N. Kelada; Praveen Sethupathy; Patrick F. Sullivan

Genome-wide association studies (GWAS) have strongly implicated MIR137 (the gene encoding the microRNA miR-137) in schizophrenia. A parsimonious hypothesis is that a pathway regulated by miR-137 is important in the etiology of schizophrenia. Full evaluation of this hypothesis requires more definitive knowledge about biological targets of miR-137, which is currently lacking. Our goals were to expand knowledge of the biology of miR-137 by identifying its empirical targets, and to test whether the resulting lists of direct and indirect targets were enriched for genes and pathways involved in risk for schizophrenia. We overexpressed miR-137 in a human neural stem cell line and analyzed gene expression changes at 24 and 48 h using RNA sequencing. Following miR-137 overexpression, 202 and 428 genes were differentially expressed after 24 and 48 h. Genes differentially expressed at 24 h were enriched for transcription factors and cell cycle genes, and differential expression at 48 h affected a wider variety of pathways. Pathways implicated in schizophrenia were upregulated in the 48 h findings (major histocompatibility complex, synapses, FMRP interacting RNAs and calcium channels). Critically, differentially expressed genes at 48 h were enriched for smaller association P-values in the largest published schizophrenia GWAS. This work provides empirical support for a role of miR-137 in the etiology of schizophrenia.


Genetics | 2014

Genetic Regulation of Zfp30, CXCL1, and Neutrophilic Inflammation in Murine Lung

Holly Rutledge; David L. Aylor; Danielle E. Carpenter; Bailey C. Peck; Peter S. Chines; Lawrence E. Ostrowski; Elissa J. Chesler; Gary A. Churchill; Fernando Pardo-Manuel de Villena; Samir N. Kelada

Allergic asthma is a complex disease characterized in part by granulocytic inflammation of the airways. In addition to eosinophils, neutrophils (PMN) are also present, particularly in cases of severe asthma. We sought to identify the genetic determinants of neutrophilic inflammation in a mouse model of house dust mite (HDM)-induced asthma. We applied an HDM model of allergic asthma to the eight founder strains of the Collaborative Cross (CC) and 151 incipient lines of the CC (preCC). Lung lavage fluid was analyzed for PMN count and the concentration of CXCL1, a hallmark PMN chemokine. PMN and CXCL1 were strongly correlated in preCC mice. We used quantitative trait locus (QTL) mapping to identify three variants affecting PMN, one of which colocalized with a QTL for CXCL1 on chromosome (Chr) 7. We used lung eQTL data to implicate a variant in the gene Zfp30 in the CXCL1/PMN response. This genetic variant regulates both CXCL1 and PMN by altering Zfp30 expression, and we model the relationships between the QTL and these three endophenotypes. We show that Zfp30 is expressed in airway epithelia in the normal mouse lung and that altering Zfp30 expression in vitro affects CXCL1 responses to an immune stimulus. Our results provide strong evidence that Zfp30 is a novel regulator of neutrophilic airway inflammation.


American Journal of Respiratory Cell and Molecular Biology | 2014

Integrative Genetic Analysis of Allergic Inflammation in the Murine Lung

Samir N. Kelada; Danielle E. Carpenter; David L. Aylor; Peter S. Chines; Holly Rutledge; Elissa J. Chesler; Gary A. Churchill; Fernando Pardo-Manuel de Villena; David A. Schwartz; Francis S. Collins

Airway allergen exposure induces inflammation among individuals with atopy that is characterized by altered airway gene expression, elevated levels of T helper type 2 cytokines, mucus hypersecretion, and airflow obstruction. To identify the genetic determinants of the airway allergen response, we employed a systems genetics approach. We applied a house dust mite mouse model of allergic airway disease to 151 incipient lines of the Collaborative Cross, a new mouse genetic reference population, and measured serum IgE, airway eosinophilia, and gene expression in the lung. Allergen-induced serum IgE and airway eosinophilia were not correlated. We detected quantitative trait loci (QTL) for airway eosinophilia on chromosome (Chr) 11 (71.802-87.098 megabases [Mb]) and allergen-induced IgE on Chr 4 (13.950-31.660 Mb). More than 4,500 genes expressed in the lung had gene expression QTL (eQTL), the majority of which were located near the gene itself. However, we also detected approximately 1,700 trans-eQTL, and many of these trans-eQTL clustered into two regions on Chr 2. We show that one of these loci (at 147.6 Mb) is associated with the expression of more than 100 genes, and, using bioinformatics resources, fine-map this locus to a 53 kb-long interval. We also use the gene expression and eQTL data to identify a candidate gene, Tlcd2, for the eosinophil QTL. Our results demonstrate that hallmark allergic airway disease phenotypes are associated with distinct genetic loci on Chrs 4 and 11, and that gene expression in the allergically inflamed lung is controlled by both cis and trans regulatory factors.


Journal of Neurology, Neurosurgery, and Psychiatry | 2003

Paraoxonase 1 promoter and coding region polymorphisms in Parkinson’s disease

Samir N. Kelada; Paola Costa-Mallen; Harvey Checkoway; Hannah Malia A Viernes; Fred M. Farin; Terri Smith-Weller; Gary M. Franklin; L. G. Costa; W. T. Longstreth; Clement E. Furlong; Gail P. Jarvik; Phillip D. Swanson

Parkinson’s disease (PD) is thought to be caused by a combination of genetic and environmental factors. Epidemiological studies have found associations of PD with pesticide exposure, or suspected pathways of pesticide exposure, such as rural residence and well water consumption. Many organophosphorous insecticides (for example, chlorpyrifos and diazinon) are bioactivated to potent cholinesterase inhibitors by the cytochromes P450, and the resulting toxic oxon forms are hydrolysed by paraoxonase (PON1). Genetic variants of detoxifying enzymes or pesticide metabolising enzymes, such as paraoxonase, may confer a predisposition to PD and thus are considered candidate genes for association studies. Multiple polymorphisms have been identified in the PON1 gene. The coding region contains two common polymorphisms, at amino acid codons 55 and 192. The glutamine (Q) to arginine (R) substitution at codon 192 causes substrate dependent differences in the kinetics of hydrolysis: compared with the PON1 192Q isoform, PON1 192R has higher activity towards paraoxon and chlorpyrifos oxon, but lower activity towards diazoxon, soman, and sarin. The leucine (L) to methionine (M) substitution at codon 55 does not affect the catalytic efficiency of substrate hydrolysis by the enzyme, but the PON1 55M allele is correlated with decreased mRNA and protein levels, because of linkage disequilibrium with a single nucleotide polymorphism (SNP) at position -108 of the promoter region of the gene. Five SNPs have been identified …

Collaboration


Dive into the Samir N. Kelada's collaboration.

Top Co-Authors

Avatar

Elissa J. Chesler

University of Tennessee Health Science Center

View shared research outputs
Top Co-Authors

Avatar

Francis S. Collins

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

David L. Aylor

North Carolina State University

View shared research outputs
Top Co-Authors

Avatar

Fernando Pardo-Manuel de Villena

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

David A. Schwartz

University of Colorado Denver

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge