Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 51
Filter
1.
Cell Genom ; 4(4): 100527, 2024 Apr 10.
Article in English | MEDLINE | ID: mdl-38537634

ABSTRACT

The seventh iteration of the reference genome assembly for Rattus norvegicus-mRatBN7.2-corrects numerous misplaced segments and reduces base-level errors by approximately 9-fold and increases contiguity by 290-fold compared with its predecessor. Gene annotations are now more complete, improving the mapping precision of genomic, transcriptomic, and proteomics datasets. We jointly analyzed 163 short-read whole-genome sequencing datasets representing 120 laboratory rat strains and substrains using mRatBN7.2. We defined ∼20.0 million sequence variations, of which 18,700 are predicted to potentially impact the function of 6,677 genes. We also generated a new rat genetic map from 1,893 heterogeneous stock rats and annotated transcription start sites and alternative polyadenylation sites. The mRatBN7.2 assembly, along with the extensive analysis of genomic variations among rat strains, enhances our understanding of the rat genome, providing researchers with an expanded resource for studies involving rats.


Subject(s)
Genome , Genomics , Rats , Animals , Genome/genetics , Molecular Sequence Annotation , Whole Genome Sequencing , Genetic Variation/genetics
2.
Am J Physiol Renal Physiol ; 325(3): F317-F327, 2023 09 01.
Article in English | MEDLINE | ID: mdl-37439198

ABSTRACT

During development of the spontaneously hypertensive rat (SHR), several distinct but closely related lines were generated. Most lines are resistant to hypertensive renal disease. However, the SHR-A3 line (stroke-prone SHR) experiences end-organ injury (EOI) and provides a model of injury susceptibility that can be used to uncover genetic causation. In the present study, we generated a congenic line in which three distinct disease loci in SHR-A3 are concurrently replaced with homologous loci from an injury-resistant SHR line (SHR-B2). Verification that all three loci were homozygously replaced in this triple congenic line [SHR-A3(Trip B2)] while the genetic background of SHR-A3 was fully retained was obtained by whole genome sequencing. Congenic genome substitution was without effect on systolic blood pressure [198.9 ± 3.34 mmHg, mean ± SE, SHR-A3(Trip B2) = 194.7 ± 2.55 mmHg]. Measures of renal injury (albuminuria, histological injury scores, and urinary biomarker levels) were reduced in SHR-A3(Trip B2) animals, even though only 4.5 Mbases of the 2.8 Gbases of the SHR-B2 genome (0.16% of the genome) was transferred into the congenic line. The gene content of the three congenic loci and the functional effects of gene polymorphism within suggest a role of immunoglobulin in EOI pathogenesis. To prove the role of antibodies in EOI in SHR-A3, we generated an SHR-A3 line in which expression from the immunoglobulin heavy chain gene was knocked out (SHR-A3-IGHKO). Animals in the SHR-A3-IGHKO line lack B cells and immunoglobulin, but the hypertensive phenotype is not affected. Renal injury, however, was reduced in this line, confirming a pathogenic role for immunoglobulin in hypertensive EOI in this model of heritable risk.NEW & NOTEWORTHY Here, we used a polygenic animal model of hypertensive renal disease to show that genetic variation affecting antibody formation underlies hypertensive renal disease. We proved the genetic thesis by generating an immunoglobulin knockout in the susceptible animal model.


Subject(s)
Hypertension , Stroke , Rats , Animals , Rats, Inbred SHR , Antibody Formation , Kidney/metabolism , Blood Pressure/genetics , Genetic Variation , Stroke/genetics , Stroke/metabolism , Stroke/pathology
3.
bioRxiv ; 2023 Sep 28.
Article in English | MEDLINE | ID: mdl-37214860

ABSTRACT

The seventh iteration of the reference genome assembly for Rattus norvegicus-mRatBN7.2-corrects numerous misplaced segments and reduces base-level errors by approximately 9-fold and increases contiguity by 290-fold compared to its predecessor. Gene annotations are now more complete, significantly improving the mapping precision of genomic, transcriptomic, and proteomics data sets. We jointly analyzed 163 short-read whole genome sequencing datasets representing 120 laboratory rat strains and substrains using mRatBN7.2. We defined ~20.0 million sequence variations, of which 18.7 thousand are predicted to potentially impact the function of 6,677 genes. We also generated a new rat genetic map from 1,893 heterogeneous stock rats and annotated transcription start sites and alternative polyadenylation sites. The mRatBN7.2 assembly, along with the extensive analysis of genomic variations among rat strains, enhances our understanding of the rat genome, providing researchers with an expanded resource for studies involving rats.

4.
Hypertension ; 80(1): 138-146, 2023 01.
Article in English | MEDLINE | ID: mdl-36330812

ABSTRACT

BACKGROUND: We report the creation and evaluation of a de novo assembly of the genome of the spontaneously hypertensive rat, the most widely used model of human cardiovascular disease. METHODS: The genome is assembled from long read sequencing (PacBio HiFi and continuous long read data [CLR]) and scaffolded with long-range structural information obtained from Bionano optical maps and proximity ligation sequencing proximity analysis of the genome. The genome assembly was polished with Illumina short reads. Completeness of the assembly was investigated using Benchmarking Universal Single Copy Orthologs analysis. The genome assembly was also evaluated with the rat reference gene set, using NCBI automated protocols. We also generated orthogonal single molecule transcript sequence reads (Iso-Seq) from 8 tissues and used them to validate the coding assembly, to annotate the assembly with RNA transcripts representing unique full length transcript isoforms for each gene and to determine whether divergences between RefSeq sequences and the assembly were attributable to assembly errors or polymorphisms. RESULTS: The assembly analysis indicates that this assembly is comparable in contiguity and completeness to the current rat reference assembly, while the use of HiFi sequencing yields an assembly that is more correct at the single base level. Synteny analysis was performed to uncover the extent of synteny and the presence and distribution of chromosomal rearrangements between the reference and this assembly. CONCLUSION: The resulting genome assembly is reference quality and captures significant structural variation.


Subject(s)
Stroke , Humans , Rats , Animals , Rats, Inbred SHR , Stroke/genetics
5.
Hypertension ; 79(10): 2239-2249, 2022 10.
Article in English | MEDLINE | ID: mdl-35950503

ABSTRACT

BACKGROUND: The spontaneously hypertensive rat (SHR) is extensively used to study hypertension. Gut microbiota dysbiosis is a notable feature in SHR for reasons unknown. Immunoglobulin A (IgA) is a major host factor required for gut microbiota homeostasis. We hypothesized that inadequate IgA contributes to gut microbiota dysbiosis in SHR. METHODS: IgA was measured in feces, cecum, serum, liver, gut-associated lymphoid tissue, and milk from SHR and Wistar Kyoto rats. IgA regulatory factors like IgM, IgG, and pIgR (polymeric immunoglobulin receptor) were analyzed. IgA and IgG antibodies and blood pressure (BP) were measured before and after administrating a bacterial antigen (ie, flagellin). RESULTS: Compared with Wistar Kyoto rats, SHR displayed remarkably near-deficient IgA levels accompanied by compensatory increases in serum IgM and IgG and gut-liver pIgR expression. Inadequate milk IgA in SHR emphasized this immune defect stemmed from the neonatal stage. Reduced IgA+ B cells in circulation and Peyer patches indicated a possible reason for the lower IgA in SHR. Noteworthy, a genetic insufficiency was unlikely because administering flagellin to SHR induced anti-flagellin IgA antibodies. This immune response surprisingly accelerated hypertension development in SHR, suggesting IgA quiescence may help maintain lower BP. CONCLUSIONS: This study is the first to reveal IgA deficiency in SHR as one host factor associated with gut microbiota dysbiosis and invigorates future research to determine the pathophysiological role of IgA in hypertension.


Subject(s)
Hypertension , IgA Deficiency , Animals , Blood Pressure , Dysbiosis , Immunoglobulin A/metabolism , Immunoglobulin G , Immunoglobulin M/metabolism , Rats , Rats, Inbred SHR , Rats, Inbred WKY
6.
Physiol Genomics ; 54(7): 251-260, 2022 07 01.
Article in English | MEDLINE | ID: mdl-35543507

ABSTRACT

Rat genomic tools have been slower to emerge than for those of humans and mice and have remained less thorough and comprehensive. The arrival of a new and improved rat reference genome, mRatBN7.2, in late 2020 is a welcome event. This assembly, like predecessor rat reference assemblies, is derived from an inbred Brown Norway rat. In this "user" survey we hope to provide other users of this assembly some insight into its characteristics and some assessment of its improvements as well as a few caveats that arise from the unique aspects of this assembly. mRatBN7.2 was generated by the Wellcome Sanger Institute as part of the large Vertebrate Genomes Project. This rat assembly has now joined human, mouse, chicken, and zebrafish in the National Center for Biotechnology Information (NCBI)'s Genome Reference Consortium, which provides ongoing curation of the assembly. Here we examine the technical procedures by which the assembly was created and assess how this assembly constitutes an improvement over its predecessor. We also indicate the technical limitations affecting the assembly, providing illustrations of how these limitations arise and the impact that results for this reference assembly.


Subject(s)
Genome , Zebrafish , Animals , Genome/genetics , Genomics/methods , Mice , Rats
7.
Hypertension ; 78(6): 1689-1700, 2021 12.
Article in English | MEDLINE | ID: mdl-34757770

ABSTRACT

The pathogenic links between elevated blood pressure and chronic kidney disease remain obscure. This article examines progress in population genetics and in animal models of hypertension and chronic kidney disease. It also provides a critique of the application of genome-wide association studies to understanding the heritability of renal function. Emerging themes identified indicate that heritable risk of chronic kidney disease in hypertension can arise from genetic variation in (1) glomerular and tubular protein handling mechanisms; (2) autoregulatory capacity of the renal vasculature; and (3) innate and adaptive immune mechanisms. Increased prevalence of hypertension-associated chronic kidney disease that occurs with aging may reflect amplification of heritable risks by normal aging processes affecting immunity and autoregulation.


Subject(s)
Hypertension/complications , Kidney/physiopathology , Renal Insufficiency, Chronic/etiology , Animals , Genomics , Humans , Hypertension/genetics , Hypertension/physiopathology , Renal Insufficiency, Chronic/genetics , Renal Insufficiency, Chronic/physiopathology
8.
Compr Physiol ; 11(4): 2433-2454, 2021 09 27.
Article in English | MEDLINE | ID: mdl-34570903

ABSTRACT

Chronic cardiovascular diseases are associated with inflammatory responses within the blood vessels and end organs. The origin of this inflammation has not been certain, and neither is its relationship to disease clear. There is a need to determine whether this association is causal or coincidental to the processes leading to cardiovascular disease. These processes are themselves complex: many cardiovascular diseases arise in conjunction with the presence of sustained elevation of blood pressure. Inflammatory processes have been linked to hypertension, and causality has been suggested. Evidence of causality poses the difficult challenge of linking the integrated and multifaceted biology of blood pressure regulation with vascular function and complex elements of immune system function. These include both, innate and adaptive immunity, as well as interactions between the host immune system and the omnipresent microorganisms that are encountered in the environment and that colonize and exist in commensal relationship with the host. Progress has been made in this task and has drawn on experimental approaches in animals, much of which have focused on hypertension occurring with prolonged infusion of angiotensin II. These laboratory studies are complemented by studies that seek to inform disease mechanism by examining the genomic basis of heritable disease susceptibility in human populations. In this realm too, evidence has emerged that implicates genetic variation affecting immunity in disease pathogenesis. In this article, we survey the genetic and genomic evidence linking high blood pressure and its end-organ injuries to immune system function and examine evidence that genomic factors can influence disease risk. © 2021 American Physiological Society. Compr Physiol 11:1-22, 2021.


Subject(s)
Cardiovascular Diseases , Adaptive Immunity , Animals , Cardiovascular Diseases/genetics , Genomics , Humans , Immunity, Innate/genetics , Inflammation/genetics
9.
Front Pharmacol ; 12: 778953, 2021.
Article in English | MEDLINE | ID: mdl-35197849

ABSTRACT

Clinical use of the combination therapy of the neprilysin inhibitor sacubitril and angiotensin II type 1 receptor blocker valsartan is known to be associated with albuminuria. Albuminuria is both a risk factor for and an indicator of kidney injury. Earlier work from our laboratory reported that the agonist of angiotensin II type 2 receptor Compound 21 (C21) prevents proteinuria, albuminuria, and is reno-protective in obese Zucker rats fed high salt diet (HSD). Thus, we hypothesized that sacubitril/C21 combination provides superior reno-protection compared to sacubitril/valsartan. Male obese Zucker rats 10-11 weeks old were treated daily with vehicle, sacubitril + C21, or sacubitril + valsartan while fed HSD for 16 days. HSD-feeding caused kidney dysfunction, evident by significant increases in urinary protein, osteopontin, and cystatin C. HSD-feeding lowered plasma cystatin C and creatinine concentrations suggestive of hyperfiltration, which was not affected by either treatment. Unlike sacubitril/valsartan, sacubitril/C21 treatment significantly decreases proteinuria, albuminuria, the expression of nephrin, and kidney weight, independent of hyperfiltration, compared with HSD alone. Moreover, sacubitril/valsartan therapy increased plasma renin and did not prevent HSD-induced increases in renal angiotensin II, while sacubitril/C21 completely prevented these changes. Together, this study suggests that sacubitril/C21 afforded superior reno-protection compared to sacubitril/valsartan therapy in high salt-fed obese Zucker rats.

10.
Genes Immun ; 21(3): 182-192, 2020 05.
Article in English | MEDLINE | ID: mdl-32300198

ABSTRACT

Similar to humans, the risk of cerebrovascular disease in stroke-prone spontaneously hypertensive rats (SHR-A3/SHRSP) arises from naturally occurring genetic variation. In the present study, we show the involvement of genetic variation affecting the store-operated calcium signaling gene, Stim1, in the pathogenesis of stroke in SHR. Stim1 is a key lymphocyte activation signaling molecule and contains functional variation in SHR-A3 that diverges from stroke-resistant SHR-B2. We created a SHR-A3 congenic line in which Stim1 was substituted with the corresponding genomic segment from SHR-B2. Compared with SHR-A3 rats, Stim1 congenic SHR-A3 (SHR-A3(Stim1-B2)) have reduced cerebrovascular disease in response to salt loading including lower neurological deficit scores and cerebral edema. Microbleeds and major hemorrhages occurred in over half of SHR-A3 rats. These lesions were absent in SHR-A3(Stim1-B2) rats. Loss of Stim1 function in mice and humans is associated with antibody-mediated autoimmunity due to defects in T lymphocyte helper function to B cells. We investigated autoantibody formation using a high-density protein array to detect the presence of IgG and IgM autoantibodies in SHR-A3. Autoantibodies to key cerebrovascular stress proteins were detected that were reduced in the congenic line.


Subject(s)
Autoantibodies/metabolism , Hypertension/genetics , Stroke/genetics , Stromal Interaction Molecule 1/genetics , Stromal Interaction Molecule 1/immunology , Animals , Animals, Congenic , Calcium/metabolism , Calcium Channels/metabolism , Calcium Signaling , Cerebrovascular Disorders/genetics , Cerebrovascular Disorders/veterinary , Disease Models, Animal , Female , Genetic Predisposition to Disease , Genetic Variation , Hypertension/complications , Hypertension/physiopathology , Male , Models, Genetic , Mutation , Rats , Rats, Inbred SHR , Stroke/complications
12.
J Am Heart Assoc ; 9(5): e014142, 2020 03 03.
Article in English | MEDLINE | ID: mdl-32075490

ABSTRACT

Background Spontaneously hypertensive rats of the stroke-prone line (SHR-A3) develop hypertensive renal disease as a result of naturally occurring genetic variation. Our prior work identified a single-nucleotide polymorphism unique to SHR-A3 that results in truncation of the carboxy terminus of STIM1. The SHR-B2 line, which is also hypertensive but resists hypertensive renal injury, expresses the wild-type STIM1. STIM1 plays a central role in lymphocyte calcium signaling that directs immune effector responses. Here we show that major defects in lymphocyte function affecting calcium signaling, nuclear factor of activated T cells activation, cytokine production, proliferation, apoptosis, and regulatory T-cell development are present in SHR-A3 and attributable to STIM1. Methods and Results To assess the role of Stim1 variation in susceptibility to hypertensive renal injury, we created a Stim1 congenic line, SHR-A3(Stim1-B2), and STIM1 function was rescued in SHR-A3. We found that Stim1 gene rescue restores disturbed lymphocyte function in SHR-A3. Hypertensive renal injury was compared in SHR-A3 and the SHR-A3(Stim1-B2) congenic line. Histologically assessed renal injury was markedly reduced in SHR-A3(Stim1-B2), as were renal injury biomarker levels measured in urine. Stim1 deficiency has been linked to the emergence of antibody-mediated autoimmunity. Renal glomerular immunoglobulin deposition was greater in SHR-A3 than SHR-B2 and was reduced by Stim1 congenic substitution. Serum anti-double-stranded DNA antibody titers in SHR-A3 were elevated compared with SHR-B2 and were reduced in SHR-A3(Stim1-B2). Conclusions Stim1 deficiency in lymphocyte function originating from Stim1 truncation in SHR-A3 combines with hypertension to create end organ disease and may do so as a result of antibody formation.


Subject(s)
CD4-Positive T-Lymphocytes/metabolism , Hypertension/complications , Kidney Diseases/etiology , Kidney/metabolism , Lymphocyte Activation , ORAI1 Protein/metabolism , Polymorphism, Single Nucleotide , Animals , Antibody-Dependent Cell Cytotoxicity , Autoimmunity , CD4-Positive T-Lymphocytes/immunology , Calcium Signaling , Cells, Cultured , Disease Models, Animal , Hypertension/genetics , Hypertension/immunology , Hypertension/metabolism , Kidney/immunology , Kidney/pathology , Kidney Diseases/genetics , Kidney Diseases/immunology , Kidney Diseases/metabolism , Male , NFATC Transcription Factors/metabolism , ORAI1 Protein/genetics , Rats, Inbred SHR , Rats, Transgenic
13.
Physiol Genomics ; 51(11): 578-585, 2019 11 01.
Article in English | MEDLINE | ID: mdl-31608789

ABSTRACT

The risk of cerebrovascular disease in stroke-prone spontaneously hypertensive rats (SHR-A3/SHRSP) arises from naturally occurring genetic variation. In the present study we show the involvement of SHR genetic variation that affects antibody formation and function in the pathogenesis of stroke. We have tested the involvement in susceptibility to stroke of genetic variation in IgH, the gene encoding the immunoglobulin heavy chain by congenic substitution. This gene contains functional natural variation in SHR-A3 that diverges from stroke-resistant SHR-B2. We created a SHR-A3 congenic line in which the IgH gene was substituted with the corresponding haplotype from SHR-B2. Compared with SHR-A3 rats, congenic substitution of the IgH locus [SHR-A3(IgH-B2)] markedly reduced cerebrovascular disease. Given the role in antibody formation of the IgH gene, we investigated the presence of IgG and IgM autoantibodies and their targets using a high-density protein array containing ~20,000 recombinant proteins. High titers of autoantibodies to key cerebrovascular stress proteins were detected, including FABP4, HSP70, and Wnt signaling proteins. Serum levels of these autoantibodies were reduced in the SHR-A3(IgH-B2) congenic line.


Subject(s)
Genetic Predisposition to Disease/genetics , Germ Cells/metabolism , Immunoglobulin Heavy Chains/genetics , Polymorphism, Single Nucleotide , Stroke/genetics , Animals , Animals, Congenic , Autoantibodies/blood , HSP70 Heat-Shock Proteins/immunology , Haplotypes , Hypertension/genetics , Immunoglobulin G/blood , Immunoglobulin M/blood , Male , Rats , Rats, Inbred SHR
14.
Hypertension ; 71(4): 700-708, 2018 04.
Article in English | MEDLINE | ID: mdl-29437896

ABSTRACT

High blood pressure exerts its deleterious effects on health largely through acceleration of end-organ diseases. Among these, progressive loss of renal function is particularly important, not only for the direct consequences of kidney damage but also because loss of renal function is associated with amplification of other adverse cardiovascular outcomes. Genetic susceptibility to hypertension and associated end-organ disease is non-Mendelian in both humans and in a rodent model, the spontaneously hypertensive rat (SHR). Here, we report that hypertensive end-organ disease in the inbred SHR-A3 line is attributable to genetic variation in the immunoglobulin heavy chain on chromosome 6. This variation coexists with variation in a 10 Mb block on chromosome 17 that contains genetic variation in 2 genes involved in immunoglobulin Fc receptor signaling. Substitution of these genomic regions into the SHR-A3 genome from the closely related, but injury-resistant, SHR-B2 line normalizes both biomarker and histological measures of renal injury. Our findings indicate that genetic variation leads to a contribution by immune mechanisms hypertensive end-organ injury and that, in this rat model, disease is influenced by differences in germ line antibody repertoire.


Subject(s)
Animals, Congenic/immunology , Chromosomes, Mammalian , Hypertension, Renal , Hypertension , Immunoglobulin Fc Fragments , Kidney , Nephritis , Receptors, Fc , Animals , Antibodies/blood , Biomarkers/blood , Chromosome Mapping , Disease Models, Animal , Genetic Predisposition to Disease , Humans , Hypertension/diagnosis , Hypertension/genetics , Hypertension, Renal/genetics , Hypertension, Renal/immunology , Hypertension, Renal/pathology , Immunoglobulin Fc Fragments/genetics , Immunoglobulin Fc Fragments/immunology , Kidney/immunology , Kidney/pathology , Nephritis/genetics , Nephritis/immunology , Nephritis/pathology , Prognosis , Quantitative Trait Loci , Rats , Rats, Inbred SHR , Receptors, Fc/genetics , Receptors, Fc/immunology
15.
J Am Heart Assoc ; 6(10)2017 Oct 05.
Article in English | MEDLINE | ID: mdl-28982675

ABSTRACT

BACKGROUND: We have investigated serum levels of immunoreactive marinobufagenin (MBG) in 16- to 20-week-old spontaneously hypertensive rats (SHRs)-A3 and in the normotensive Wistar-Kyoto (WKY) rat strain in the absence of salt loading, and we have investigated the genetic control of serum MBG. METHODS AND RESULTS: We genotyped the F2 progeny of an SHR-A3×WKY intercross using a genome-wide panel of 253 single-nucleotide polymorphism markers that were dimorphic between SHR-A3 and WKY and measured serum MBG by ELISA. Serum MBG levels were lower in SHR-A3 than WKY rats (0.39±0.07 and 1.27±0.40 nmol/L, respectively), suggesting that MBG may not play a role in the markedly divergent blood pressure measured by telemetry in rats of these 2 strains (SHR-A3 and WKY, 198.3±4.43 and 116.8±1.51 mm Hg, respectively). The strain difference in serum MBG was investigated to determine whether genomic regions influencing MBG might be identified by genetic mapping. Quantitative trait locus mapping indicated a single locus influencing serum MBG in the region of chromosome 6q12. Homozygosity of WKY alleles at this locus was associated with increased serum MBG levels. We surveyed whole genome sequences from our SHR-A3 and WKY lines, seeking coding sequence variation between SHR-A3 and WKY within the mapped locus that might explain the inherited strain difference in serum MBG. CONCLUSIONS: We identified amino acid substitution in the sterol transport protein Abcg5, present in SHR-A3, but absent in WKY, that is a potential mechanism influencing MBG levels.


Subject(s)
ATP Binding Cassette Transporter, Subfamily G, Member 5/genetics , Blood Pressure/genetics , Bufanolides/blood , Hypertension/genetics , Lipoproteins/genetics , Polymorphism, Single Nucleotide , Animals , Biomarkers/blood , Crosses, Genetic , Disease Models, Animal , Genetic Predisposition to Disease , Genome-Wide Association Study , Homozygote , Hypertension/blood , Hypertension/physiopathology , Male , Phenotype , Quantitative Trait Loci , Rats, Inbred SHR , Rats, Inbred WKY
16.
Physiol Genomics ; 49(11): 601-617, 2017 Nov 01.
Article in English | MEDLINE | ID: mdl-28916635

ABSTRACT

The application of gene mapping methods to uncover the genetic basis of hypertension in the inbred spontaneously hypertensive rat (SHR) began over 25 yr ago. This animal provides a useful model of genetic high blood pressure, and some of its features are described. In particular, it appears to be a polygenic model of disease, and polygenes participate in human hypertension genetic risk. The SHR hypertension alleles were fixed rapidly by selective breeding in just a few generations and so are presumably common genetic variants present in the outbred Wistar strain from which SHR was created. This review provides a background to the origins and genesis of this rat line. It considers its usefulness as a model organism for a common cardiovascular disease. The progress and obstacles facing mapping are considered in depth, as are the emergence and application of other genome-wide genetic discovery approaches that have been applied to investigate this model. Candidate genes, their identification, and the evidence to support their potential role in blood pressure elevation are considered. The review assesses the progress that has arisen from this work has been limited. Consideration is given to some of the factors that have impeded progress, and prospects for advancing understanding of the genetic basis of hypertension in this model are discussed.


Subject(s)
Genetic Predisposition to Disease , Hypertension/genetics , Animals , Blood Pressure/genetics , Chromosome Mapping , Genetic Loci , Humans , Hypertension/physiopathology , Rats, Inbred SHR
17.
Am J Physiol Renal Physiol ; 312(5): F861-F869, 2017 05 01.
Article in English | MEDLINE | ID: mdl-28228406

ABSTRACT

Lymphatic vessels are vital for the trafficking of immune cells from the interstitium to draining lymph nodes during inflammation. Hypertension is associated with renal infiltration of activated immune cells and inflammation; however, it is unknown how renal lymphatic vessels change in hypertension. We hypothesized that renal macrophage infiltration and inflammation would cause increased lymphatic vessel density in hypertensive rats. Spontaneously hypertensive rats (SHR) that exhibit hypertension and renal injury (SHR-A3 strain) had significantly increased renal lymphatic vessel density and macrophages at 40 wk of age compared with Wistar-Kyoto (WKY) controls. SHR rats that exhibit hypertension but minimal renal injury (SHR-B2 strain) had significantly less renal lymphatic vessel density compared with WKY rats. The signals for lymphangiogenesis, VEGF-C and its receptor VEGF-R3, and proinflammatory cytokine genes increased significantly in the kidneys of SHR-A3 rats but not in SHR-B2 rats. Fischer 344 rats exhibit normal blood pressure but develop renal injury as they age. Kidneys from 24-mo- and/or 20-mo-old Fischer rats had significantly increased lymphatic vessel density, macrophage infiltration, VEGF-C and VEGF-R3 expression, and proinflammatory cytokine gene expression compared with 4-mo-old controls. These data together demonstrate that renal immune cell infiltration and inflammation cause lymphangiogenesis in hypertension- and aging-associated renal injury.


Subject(s)
Blood Pressure , Hypertension/complications , Kidney/physiopathology , Lymphangiogenesis , Lymphatic Vessels/physiopathology , Nephritis/etiology , Age Factors , Animals , Cytokines/metabolism , Disease Models, Animal , Hypertension/pathology , Hypertension/physiopathology , Inflammation Mediators/metabolism , Kidney/immunology , Kidney/metabolism , Kidney/pathology , Lymphatic Vessels/immunology , Lymphatic Vessels/metabolism , Lymphatic Vessels/pathology , Macrophages/immunology , Macrophages/metabolism , Macrophages/pathology , Nephritis/immunology , Nephritis/pathology , Nephritis/physiopathology , Rats, Inbred F344 , Rats, Inbred SHR , Rats, Inbred WKY , Species Specificity , Vascular Endothelial Growth Factor C/metabolism , Vascular Endothelial Growth Factor Receptor-3/metabolism
18.
Physiol Genomics ; 49(3): 132-140, 2017 03 01.
Article in English | MEDLINE | ID: mdl-28011882

ABSTRACT

Stroke-prone spontaneously hypertensive rats (SHR-A3) develop strokes and progressive kidney disease as a result of naturally occurring genetic variations. We recently identified genetic variants in immune signaling pathways that contribute to end-organ injury. The present study was designed to test the hypothesis that a dysregulated immune response promotes stroke susceptibility. We salt-loaded 20 wk old male SHR-A3 rats and treated them with the immunosuppressant mycophenolate mofetil (MMF, 25 mg/kg/day po) (n = 8) or vehicle (saline) (n = 9) for 8 wk. Blood pressure (BP) was measured weekly by telemetry. Compared with vehicle-treated controls, MMF-treated SHR-A3 rats had improved survival and lower neurological deficit scores (1.44 vs. 0.125; P < 0.02). Gross morphology of the brain revealed cerebral edema in 8 of 9, and microbleeds and hemorrhages in 5 of 9 vehicle-treated rats. These lesions were absent in MMF-treated rats. Brain CD68 expression, indicating macrophage/microglial activation, was upregulated in vehicle-treated rats with microbleeds and hemorrhages but was undetectable in the brains of MMF-treated rats. MMF also prevented renal injury in SHR-A3 rats, evidenced by reduced proteinuria (albumin:creatinine) from 7.52 to 1.05 mg/mg (P < 0.03) and lower tubulointerstitial injury scores (2.46 vs. 1.43; P < 0.01). Salt loading resulted in a progressive increase in BP, which was blunted in rats receiving MMF. Our findings provide evidence that abnormal immune activation predisposes to cerebrovascular and renal injury in stroke-prone SHR-A3 rats.


Subject(s)
Brain Injuries/etiology , Brain Injuries/prevention & control , Mycophenolic Acid/therapeutic use , Stroke/complications , Stroke/drug therapy , Animals , Blood Pressure , Brain Edema/complications , Brain Edema/pathology , Brain Edema/physiopathology , Brain Injuries/physiopathology , Immunosuppression Therapy , Inflammation/pathology , Kidney/injuries , Kidney/pathology , Kidney/physiopathology , Mycophenolic Acid/pharmacology , Rats, Inbred SHR , Stroke/physiopathology , Survival Analysis
19.
J Am Soc Nephrol ; 27(7): 2035-48, 2016 07.
Article in English | MEDLINE | ID: mdl-26574044

ABSTRACT

Store-operated calcium entry (SOCE) is the mechanism by which extracellular signals elicit prolonged intracellular calcium elevation to drive changes in fundamental cellular processes. Here, we investigated the role of SOCE in the regulation of renal water reabsorption, using the inbred rat strain SHR-A3 as an animal model with disrupted SOCE. We found that SHR-A3, but not SHR-B2, have a novel truncating mutation in the gene encoding stromal interaction molecule 1 (STIM1), the endoplasmic reticulum calcium (Ca(2+)) sensor that triggers SOCE. Balance studies revealed increased urine volume, hypertonic plasma, polydipsia, and impaired urinary concentrating ability accompanied by elevated circulating arginine vasopressin (AVP) levels in SHR-A3 compared with SHR-B2. Isolated, split-open collecting ducts (CD) from SHR-A3 displayed decreased basal intracellular Ca(2+) levels and a major defect in SOCE. Consequently, AVP failed to induce the sustained intracellular Ca(2+) mobilization that requires SOCE in CD cells from SHR-A3. This effect decreased the abundance of aquaporin 2 and enhanced its intracellular retention, suggesting impaired sensitivity of the CD to AVP in SHR-A3. Stim1 knockdown in cultured mpkCCDc14 cells reduced SOCE and basal intracellular Ca(2+) levels and prevented AVP-induced translocation of aquaporin 2, further suggesting the effects in SHR-A3 result from the expression of truncated STIM1. Overall, these results identify a novel mechanism of nephrogenic diabetes insipidus and uncover a role of SOCE in renal water handling.


Subject(s)
Calcium Channels/metabolism , Diabetes Insipidus, Nephrogenic/etiology , Diabetes Insipidus, Nephrogenic/metabolism , Animals , Aquaporin 2/physiology , Arginine Vasopressin/physiology , Cells, Cultured , Male , Rats , Rats, Inbred SHR/genetics , Stromal Interaction Molecule 1/physiology
20.
Circ Cardiovasc Genet ; 7(6): 903-10, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25366137

ABSTRACT

BACKGROUND: The spontaneously hypertensive rat (SHR) strain exists in lines that contrast strongly in susceptibility to renal injury in hypertension. These inbred lines share common ancestry, and only 13% of their genomes arise from different ancestors. METHODS AND RESULTS: We used next gen sequencing to detect natural allelic variation in 5 genes of the immunoreceptor signaling pathway (IgH, Dok3, Src, Syk, and JunD) that arise from different ancestors in the injury-prone SHR-A3 and the resistant SHR-B2 lines. We created an intercross between these lines, and in the F2 progeny, we observed that the inheritance of haplotype blocks containing the SHR-A3 alleles of these 5 genes correlated with increased albuminuria and histological measures of renal injury. To test whether accumulated genetic variation in this pathway may create a therapeutic target in hypertensive renal injury, rats of both lines were treated with the immunosuppressant mycophenolate mofetil (MMF). MMF reduced proteinuria (albumin to creatinine ratio) from 6.6 to 1.2 mg/mg (P<0.001) in SHR-A3. Glomerular injury scores were reduced in MMF-treated SHR-A3 from 1.6 to 1.4 (P<0.002). Tubulo-interstitial injury was reduced in MMF-treated SHR-A3 from 2.62 to 2.0 (P=0.001). MMF treatment also reduced renal fibrosis in SHR-A3 (3.9 versus 2.0; P<0.001). CONCLUSIONS: Polygenic susceptibility to renal injury in hypertension arises in association with genetic variation in genes that participate in immune responses and is dramatically improved by reduction of immune system activity.


Subject(s)
Genetic Variation , Kidney Diseases/genetics , Receptors, Immunologic/genetics , Signal Transduction , Albuminuria/drug therapy , Albuminuria/etiology , Alleles , Animals , Blood Pressure , Genetic Predisposition to Disease , Glomerular Filtration Rate , Haplotypes , Hypertension/complications , Immunosuppressive Agents/therapeutic use , Kidney/pathology , Kidney Diseases/drug therapy , Kidney Diseases/etiology , Lymphocytes/cytology , Lymphocytes/immunology , Mycophenolic Acid/analogs & derivatives , Mycophenolic Acid/therapeutic use , Rats , Rats, Inbred SHR , Receptors, Immunologic/metabolism , Sequence Analysis, DNA
SELECTION OF CITATIONS
SEARCH DETAIL
...