Network


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

Hotspot


Dive into the research topics where Robert H. Wallis is active.

Publication


Featured researches published by Robert H. Wallis.


Nature Genetics | 2007

Direct quantitative trait locus mapping of mammalian metabolic phenotypes in diabetic and normoglycemic rat models

Marc-Emmanuel Dumas; Steven P. Wilder; Marie-Thérèse Bihoreau; Richard H. Barton; Jane Fearnside; Karène Argoud; Lisa D'Amato; Robert H. Wallis; Christine Blancher; Hector C. Keun; Dorrit Baunsgaard; James Scott; Ulla G. Sidelmann; Jeremy K. Nicholson; Dominique Gauguier

Characterizing the relationships between genomic and phenotypic variation is essential to understanding disease etiology. Information-dense data sets derived from pathophysiological, proteomic and transcriptomic profiling have been applied to map quantitative trait loci (QTLs). Metabolic traits, already used in QTL studies in plants, are essential phenotypes in mammalian genetics to define disease biomarkers. Using a complex mammalian system, here we show chromosomal mapping of untargeted plasma metabolic fingerprints derived from NMR spectroscopic analysis in a cross between diabetic and control rats. We propose candidate metabolites for the most significant QTLs. Metabolite profiling in congenic strains provided evidence of QTL replication. Linkage to a gut microbial metabolite (benzoate) can be explained by deletion of a uridine diphosphate glucuronosyltransferase. Mapping metabotypic QTLs provides a practical approach to understanding genome-phenotype relationships in mammals and may uncover deeper biological complexity, as extended genome (microbiome) perturbations that affect disease processes through transgenomic effects may influence QTL detection.


Hypertension | 1996

Analysis of Quantitative Trait Loci for Blood Pressure on Rat Chromosomes 2 and 13 Age-Related Differences in Effect

Nilesh J. Samani; Dominique Gauguier; Madeleine Vincent; Michael A. Kaiser; Marie-Thérèse Bihoreau; David Lodwick; Robert H. Wallis; Valerie Parent; Phillip Kimber; Fraser Rattray; John R. Thompson; Jean Sassard; Mark Lathrop

Previous studies have suggested the presence of quantitative trait loci (QTLs) influencing blood pressure on rat chromosomes 2 and 13. In this study, we mapped the QTLs in F2 rats derived from a cross of the spontaneously hypertensive rat and the Wistar-Kyoto rat and analyzed the effect of the QTLs on blood pressures measured longitudinally between 12 and 25 weeks of age. We analyzed 16 polymorphic markers spanning 147.3 cM on chromosome 2 and 13 markers spanning 91.6 cM on chromosome 13. Both chromosomes contained QTLs with highly significant effects on blood pressure (peak logarithm of the odds [LOD] scores, 5.64 and 5.75, respectively). On chromosome 2, the peak was localized to a position at anonymous marker D2Wox7, 2.9 cM away from the gene for the sodium-potassium ATPase alpha 1-subunit. On chromosome 13, the major peak coincided with the marker D13Mit2, 21.7 cM away from the renin gene, but there was a suggestion of multiple peaks. The effect of the QTL on chromosome 2 was seen throughout from 12 to 25 weeks of age, whereas interestingly, the effect for the QTL on chromosome 13 was maximal at 20 weeks of age but disappeared at 25 weeks of age, presumably because of the effect of either epistatic factors or environmental influences. The findings provide important information on QTLs influencing blood pressure on rat chromosomes 2 and 13 that will be useful in localizing and identifying the causative genes and emphasize the importance of age being taken into account when the effects of individual QTLs on a trait that shows significant age-related changes are being analyzed.


Mammalian Genome | 1999

A gene map of the rat derived from linkage analysis and related regions in the mouse and human genomes.

Dominique Gauguier; Pamela J. Kaisaki; Mathias Rouard; Robert H. Wallis; Julie Browne; John P. Rapp; Marie-Thérèse Bihoreau

Abstract. We report the localization by linkage analysis in the rat genome of 148 new markers derived from 128 distinct known gene sequences, ESTs, and anonymous sequences selected in GenBank database on the basis of the presence of a repeated element. The composite linkage map of the rat contributed by our group integrates mapping information on a total of 370 different known genes, ESTs, and anonymous mouse or human sequences, and provides a valuable tool for comparative genome analysis. 206 and 254 homologous loci were identified in the mouse and human genomes respectively. Our linkage map, which combines both anonymous markers and gene markers, should facilitate the advancement of genetic studies for a wide variety of rat models characterized for complete phenotypes. The comparative genome mapping should define genetic regions in human likely to be homologous to susceptibility loci identified in rat and provide useful information for the identification of new potential candidates for genetic disorders.


Diabetologia | 2004

Enhanced insulin secretion and cholesterol metabolism in congenic strains of the spontaneously diabetic (Type 2) Goto Kakizaki rat are controlled by independent genetic loci in rat chromosome 8

Robert H. Wallis; Karin J. Wallace; Stephan C. Collins; M. McAteer; Karène Argoud; M. T. Bihoreau; Pamela J. Kaisaki; Dominique Gauguier

Aims/hypothesisGenetic investigations in the spontaneously diabetic (Type 2) Goto Kakizaki (GK) rat have identified quantitative trait loci (QTL) for diabetes-related phenotypes. The aims of this study were to refine the chromosomal mapping of a QTL (Nidd/gk5) identified in chromosome 8 of the GK rat and to define a pathophysiological profile of GK gene variants underlying the QTL effects in congenics.MethodsGenetic linkage analysis was carried out with chromosome 8 markers genotyped in a GKxBN F2 intercross previously used to map diabetes QTL. Two congenic strains were designed to contain GK haplotypes in the region of Nidd/gk5 transferred onto a Brown Norway (BN) genetic background, and a broad spectrum of diabetes phenotypes were characterised in the animals.ResultsResults from QTL mapping suggest that variations in glucose-stimulated insulin secretion in vivo, and in body weight are controlled by different chromosome 8 loci (LOD3.53; p=0.0004 and LOD4.19; p=0.00007, respectively). Extensive physiological screening in male and female congenics at 12 and 24 weeks revealed the existence of GK variants at the locus Nidd/gk5, independently responsible for significantly enhanced insulin secretion and increased levels of plasma triglycerides, phospholipids and HDL, LDL and total cholesterol. Sequence polymorphisms detected between the BN and GK strains in genes encoding ApoAI, AIV, CIII and Lipc do not account for these effects.Conclusions/interpretationWe refined the localisation of the QTL Nidd/gk5 and its pathophysiological characteristics in congenic strains derived for the locus. These congenic strains provide novel models for testing the contribution of a subset of GK alleles on diabetes phenotypes and for identifying diabetes susceptibility genes.


PLOS ONE | 2008

Pathophysiological, genetic and gene expression features of a novel rodent model of the cardio-metabolic syndrome.

Robert H. Wallis; Stephan C. Collins; Pamela J. Kaisaki; Karène Argoud; Steven P. Wilder; Karin J. Wallace; Massimiliano Ria; Alain Ktorza; Patrik Rorsman; Marie-Thérèse Bihoreau; Dominique Gauguier

Background Complex etiology and pathogenesis of pathophysiological components of the cardio-metabolic syndrome have been demonstrated in humans and animal models. Methodology/Principal Findings We have generated extensive physiological, genetic and genome-wide gene expression profiles in a congenic strain of the spontaneously diabetic Goto-Kakizaki (GK) rat containing a large region (110 cM, 170 Mb) of rat chromosome 1 (RNO1), which covers diabetes and obesity quantitative trait loci (QTL), introgressed onto the genetic background of the normoglycaemic Brown Norway (BN) strain. This novel disease model, which by the length of the congenic region closely mirrors the situation of a chromosome substitution strain, exhibits a wide range of abnormalities directly relevant to components of the cardio-metabolic syndrome and diabetes complications, including hyperglycaemia, hyperinsulinaemia, enhanced insulin secretion both in vivo and in vitro, insulin resistance, hypertriglyceridemia and altered pancreatic and renal histological structures. Gene transcription data in kidney, liver, skeletal muscle and white adipose tissue indicate that a disproportionately high number (43–83%) of genes differentially expressed between congenic and BN rats map to the GK genomic interval targeted in the congenic strain, which represents less than 5% of the total length of the rat genome. Genotype analysis of single nucleotide polymorphisms (SNPs) in strains genetically related to the GK highlights clusters of conserved and strain-specific variants in RNO1 that can assist the identification of naturally occurring variants isolated in diabetic and hypertensive strains when different phenotype selection procedures were applied. Conclusions Our results emphasize the importance of rat congenic models for defining the impact of genetic variants in well-characterised QTL regions on in vivo pathophysiological features and cis-/trans- regulation of gene expression. The congenic strain reported here provides a novel and sustainable model for investigating the pathogenesis and genetic basis of risks factors for the cardio-metabolic syndrome.


Mammalian Genome | 2003

Marker-assisted congenic screening (MACS): a database tool for the efficient production and characterization of congenic lines.

Stephan C. Collins; Robert H. Wallis; Karin J. Wallace; Mt Bihoreau; Dominique Gauguier

Over the past decades, genetic studies in rodent models of human multifactorial disorders have led to the detection of numerous chromosomal regions associated with disease phenotypes. Owing to the complex control of these phenotypes and the size of the disease loci, identifying the underlying genes requires further analyses in new original models, including chromosome substitution (consomic) and congenic lines, derived to evaluate the phenotypic effects of disease susceptibility loci and fine-map the disease genes. We have developed a relational database (MACS) specifically designed for the genetic marker-assisted production of large series of rodent consomic and congenic lines (“speed congenics”), the organization of their genetic and phenotypic characterizations, and the acquisition and archiving of both genetic and phenotypic data. This database, originally optimized for the production of rat congenics, can also be applied to mouse mapping projects. MACS represents an essential system for significantly improving efficiency and accuracy in investigations of multiple consomic and congenic lines simultaneously derived for different disease loci, and ultimately cloning genes underlying complex phenotypes.


Mammalian Genome | 1998

Localization of tub and uncoupling proteins (Ucp) 2 and 3 to a region of rat Chromosome 1 linked to glucose intolerance and adiposity in the Goto-Kakizaki (GK) Type 2 diabetic rat

Pamela J. Kaisaki; Peng Y. Woon; Robert H. Wallis; Anthony P. Monaco; M Lathrop; Dominique Gauguier

Uncoupling proteins (UCP) 2 and UCP3 are closely related to themitochondrial membrane protein UCP1, which can dissociate res-piration from ATP synthesis, resulting in the generation of heat(Fleury et al. 1997; Boss et al. 1997; Vidal-Puig et al. 1997).Because this property may allow for disposal of excess energy,uncoupling proteins are regarded as potential candidate genes forobesity and Type 2 diabetes. Another candidate gene for obesity,tub, was identified as an autosomal recessive mutation causingobesity in mouse (Noben-Trauth et al. 1996; Kleyn et al. 1996).Tub, Ucp2, and Ucp3 have been mapped to a region of mouseChromosome (Chr) 7 that shares synteny homology with humanChr 11p15-11q13 (Chung et al. 1996; Noben-Trauth et al. 1996;Kleyn et al. 1996; Fleury et al. 1997; Solanes et al. 1997). Ucp3has been shown in humans and mice to reside within 75–150 kbof Ucp2, co-localizing in P1 and BAC genomic clones (Solanes etal. 1997). Ucp2 and 3 are located at quantitative trait loci (QTL)for obesity in three different mouse models (Seldin et al. 1994;Warden et al. 1995; Taylor and Phillips 1996) and show linkagewith resting metabolic rate in humans (Bouchard et al. 1997). RatcDNAs for Ucp2 (Matsuda et al. 1997; GenBank AB006613) andUcp3 (GenBank U92069) have been cloned, but these genes havenot yet been mapped in rat. Tub has not yet been cloned or mappedin rat.A segment of rat Chr 1 that shares synteny homology withmouse Chr 7 holds areas of interest for several diseases. Includedare regions linked to glucose intolerance and adiposity in the Goto-Kakizaki (GK) nonobese Type 2 diabetic rat (Gauguier et al. 1996;Galli et al. 1996), hypertension in several rat strains (spontane-ously hypertensive, stroke-prone spontaneously hypertensive, re-combinant inbred strains, and Dahl salt-sensitive hypertensive; re-viewed in Jacob et al. 1996), hypertension and renal failure in thefawn-hooded rat (Brown et al. 1996), pancreatic islet morphologyin obese WKY13M F2 Lepr


Diabetologia | 2006

Genetic control of plasma lipid levels in a cross derived from normoglycaemic Brown Norway and spontaneously diabetic Goto-Kakizaki rats.

Karène Argoud; Steven P. Wilder; M. McAteer; M. T. Bihoreau; F. Ouali; P. Y. Woon; Robert H. Wallis; Alain Ktorza; Dominique Gauguier

Aims/hypothesisDyslipidaemia is a main component of the insulin resistance syndrome. The inbred Goto–Kakizaki (GK) rat is a model of spontaneous type 2 diabetes and insulin resistance, which has been used to identify diabetes-related susceptibility loci in genetic crosses. The objective of our study was to test the genetic control of lipid metabolism in the GK rat and investigate a possible relationship with known genetic loci regulating glucose homeostasis in this strain.Materials and methodsPlasma concentration of triglycerides, phospholipids, total cholesterol, HDL, LDL and VLDL cholesterol were determined in a cohort of 151 hybrids of an F2 cross derived from GK and non-diabetic Brown Norway (BN) rats. Data from the genome-wide scan of the F2 hybrids were used to test for evidence of genetic linkage to the lipid quantitative traits.ResultsWe identified statistically significant quantitative trait loci (QTLs) that control the level of plasma phospholipids and triglycerides (chromosome 1), LDL cholesterol (chromosome 3) and total and HDL cholesterol (chromosomes 1 and 5). These QTLs do not coincide with previously identified diabetes susceptibility loci in a similar cross. The significance of lipid QTLs mapped to chromosomes 1 and 5 is strongly influenced by sex.Conclusion/interpretationWe established that several genetic loci control the quantitative variations of plasma lipid variables in a GK×BN cross. They appear to be distinct from known GK diabetes QTLs, indicating that lipid metabolism and traits directly relevant to glucose and insulin regulation are controlled by different gene variants in this strain combination.


Mammalian Genome | 2006

Mapping diabetes QTL in an intercross derived from a congenic strain of the Brown Norway and Goto-Kakizaki rats

Stephan C. Collins; Robert H. Wallis; Steven P. Wilder; Karin J. Wallace; Karène Argoud; Pamela J. Kaisaki; Marie-Thérèse Bihoreau; Dominique Gauguier

Genetic studies in experimental crosses derived from the inbred Goto-Kakizaki (GK) rat model of spontaneous diabetes mellitus have identified quantitative trait loci (QTL) for diabetes phenotypes in a large region of rat Chromosome (RNO) 1. To test the impact of GK variants on QTL statistical and biological features, we combined genetic and physiologic studies in a cohort of F2 hybrids derived from a QTL substitution congenic strain (QTLSCS) carrying a 110-cM GK haplotype of RNO1 introgressed onto the genetic background of the Brown Norway (BN) strain. Glucose intolerance and altered insulin secretion in QTLSCS rats when compared with BN controls were consistent with original QTL features in a GK × BN F2 cross. Segregating GK alleles in the QTLSCS F2 cross account for most of these phenotypic differences between QTLSCS and BN rats. However, significant QTL for diabetes traits in both the QTLSCS and GK × BN F2 cohorts account for a similar small proportion of their variance. Comparing results from these experimental systems provides indirect estimates of the contribution of genetic interactions and environmental factors to QTL architecture as well as locus and biological targets for future post-QTL mapping studies in congenic substrains.


G3: Genes, Genomes, Genetics | 2016

Transcriptome Profiling in Rat Inbred Strains and Experimental Cross Reveals Discrepant Genetic Architecture of Genome-Wide Gene Expression

Pamela J. Kaisaki; Georg W. Otto; Karène Argoud; Stephan C. Collins; Robert H. Wallis; Steven P. Wilder; Anthony C.Y. Yau; Christophe Hue; S. Calderari; Marie-Thérèse Bihoreau; Jean-Baptiste Cazier; Richard Mott; Dominique Gauguier

To test the impact of genetic heterogeneity on cis- and trans-mediated mechanisms of gene expression regulation, we profiled the transcriptome of adipose tissue in 20 inbred congenic strains derived from diabetic Goto–Kakizaki (GK) rats and Brown–Norway (BN) controls, which contain well-defined blocks (1–183 Mb) of genetic polymorphisms, and in 123 genetically heterogeneous rats of an (GK × BN)F2 offspring. Within each congenic we identified 73–1351 differentially expressed genes (DEGs), only 7.7% of which mapped within the congenic blocks, and which may be regulated in cis. The remainder localized outside the blocks, and therefore must be regulated in trans. Most trans-regulated genes exhibited approximately twofold expression changes, consistent with monoallelic expression. Altered biological pathways were replicated between congenic strains sharing blocks of genetic polymorphisms, but polymorphisms at different loci also had redundant effects on transcription of common distant genes and pathways. We mapped 2735 expression quantitative trait loci (eQTL) in the F2 cross, including 26% predominantly cis-regulated genes, which validated DEGs in congenic strains. A hotspot of >300 eQTL in a 10 cM region of chromosome 1 was enriched in DEGs in a congenic strain. However, many DEGs among GK, BN and congenic strains did not replicate as eQTL in F2 hybrids, demonstrating distinct mechanisms of gene expression when alleles segregate in an outbred population or are fixed homozygous across the entire genome or in short genomic regions. Our analysis provides conceptual advances in our understanding of the complex architecture of genome expression and pathway regulation, and suggests a prominent impact of epistasis and monoallelic expression on gene transcription.

Collaboration


Dive into the Robert H. Wallis's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Karène Argoud

Wellcome Trust Centre for Human Genetics

View shared research outputs
Top Co-Authors

Avatar

Marie-Thérèse Bihoreau

Wellcome Trust Centre for Human Genetics

View shared research outputs
Top Co-Authors

Avatar

Pamela J. Kaisaki

Wellcome Trust Centre for Human Genetics

View shared research outputs
Top Co-Authors

Avatar

Steven P. Wilder

Wellcome Trust Centre for Human Genetics

View shared research outputs
Top Co-Authors

Avatar

Karin J. Wallace

Wellcome Trust Centre for Human Genetics

View shared research outputs
Top Co-Authors

Avatar

Georg W. Otto

Wellcome Trust Centre for Human Genetics

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge