Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Am J Physiol Renal Physiol ; 325(3): F317-F327, 2023 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-37439198

RESUMEN

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.


Asunto(s)
Hipertensión , Accidente Cerebrovascular , Ratas , Animales , Ratas Endogámicas SHR , Formación de Anticuerpos , Riñón/metabolismo , Presión Sanguínea/genética , Variación Genética , Accidente Cerebrovascular/genética , Accidente Cerebrovascular/metabolismo , Accidente Cerebrovascular/patología
2.
Physiol Genomics ; 54(7): 251-260, 2022 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-35543507

RESUMEN

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.


Asunto(s)
Genoma , Pez Cebra , Animales , Genoma/genética , Genómica/métodos , Ratones , Ratas
3.
Genes Immun ; 21(3): 182-192, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32300198

RESUMEN

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.


Asunto(s)
Autoanticuerpos/metabolismo , Hipertensión/genética , Accidente Cerebrovascular/genética , Molécula de Interacción Estromal 1/genética , Molécula de Interacción Estromal 1/inmunología , Animales , Animales Congénicos , Calcio/metabolismo , Canales de Calcio/metabolismo , Señalización del Calcio , Trastornos Cerebrovasculares/genética , Trastornos Cerebrovasculares/veterinaria , Modelos Animales de Enfermedad , Femenino , Predisposición Genética a la Enfermedad , Variación Genética , Hipertensión/complicaciones , Hipertensión/fisiopatología , Masculino , Modelos Genéticos , Mutación , Ratas , Ratas Endogámicas SHR , Accidente Cerebrovascular/complicaciones
4.
Physiol Genomics ; 51(11): 578-585, 2019 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-31608789

RESUMEN

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.


Asunto(s)
Predisposición Genética a la Enfermedad/genética , Células Germinativas/metabolismo , Cadenas Pesadas de Inmunoglobulina/genética , Polimorfismo de Nucleótido Simple , Accidente Cerebrovascular/genética , Animales , Animales Congénicos , Autoanticuerpos/sangre , Proteínas HSP70 de Choque Térmico/inmunología , Haplotipos , Hipertensión/genética , Inmunoglobulina G/sangre , Inmunoglobulina M/sangre , Masculino , Ratas , Ratas Endogámicas SHR
5.
Physiol Genomics ; 49(3): 132-140, 2017 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-28011882

RESUMEN

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.


Asunto(s)
Lesiones Encefálicas/etiología , Lesiones Encefálicas/prevención & control , Ácido Micofenólico/uso terapéutico , Accidente Cerebrovascular/complicaciones , Accidente Cerebrovascular/tratamiento farmacológico , Animales , Presión Sanguínea , Edema Encefálico/complicaciones , Edema Encefálico/patología , Edema Encefálico/fisiopatología , Lesiones Encefálicas/fisiopatología , Terapia de Inmunosupresión , Inflamación/patología , Riñón/lesiones , Riñón/patología , Riñón/fisiopatología , Ácido Micofenólico/farmacología , Ratas Endogámicas SHR , Accidente Cerebrovascular/fisiopatología , Análisis de Supervivencia
7.
Hypertension ; 80(1): 138-146, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36330812

RESUMEN

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.


Asunto(s)
Accidente Cerebrovascular , Humanos , Ratas , Animales , Ratas Endogámicas SHR , Accidente Cerebrovascular/genética
8.
Compr Physiol ; 11(4): 2433-2454, 2021 09 27.
Artículo en Inglés | MEDLINE | ID: mdl-34570903

RESUMEN

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.


Asunto(s)
Enfermedades Cardiovasculares , Inmunidad Adaptativa , Animales , Enfermedades Cardiovasculares/genética , Genómica , Humanos , Inmunidad Innata/genética , Inflamación/genética
9.
Hypertension ; 78(6): 1689-1700, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34757770

RESUMEN

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.


Asunto(s)
Hipertensión/complicaciones , Riñón/fisiopatología , Insuficiencia Renal Crónica/etiología , Animales , Genómica , Humanos , Hipertensión/genética , Hipertensión/fisiopatología , Insuficiencia Renal Crónica/genética , Insuficiencia Renal Crónica/fisiopatología
10.
J Am Heart Assoc ; 9(5): e014142, 2020 03 03.
Artículo en Inglés | MEDLINE | ID: mdl-32075490

RESUMEN

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.


Asunto(s)
Linfocitos T CD4-Positivos/metabolismo , Hipertensión/complicaciones , Enfermedades Renales/etiología , Riñón/metabolismo , Activación de Linfocitos , Proteína ORAI1/metabolismo , Polimorfismo de Nucleótido Simple , Animales , Citotoxicidad Celular Dependiente de Anticuerpos , Autoinmunidad , Linfocitos T CD4-Positivos/inmunología , Señalización del Calcio , Células Cultivadas , Modelos Animales de Enfermedad , Hipertensión/genética , Hipertensión/inmunología , Hipertensión/metabolismo , Riñón/inmunología , Riñón/patología , Enfermedades Renales/genética , Enfermedades Renales/inmunología , Enfermedades Renales/metabolismo , Masculino , Factores de Transcripción NFATC/metabolismo , Proteína ORAI1/genética , Ratas Endogámicas SHR , Ratas Transgénicas
11.
Hypertension ; 71(4): 700-708, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29437896

RESUMEN

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.


Asunto(s)
Animales Congénicos/inmunología , Cromosomas de los Mamíferos , Hipertensión Renal , Hipertensión , Fragmentos Fc de Inmunoglobulinas , Riñón , Nefritis , Receptores Fc , Animales , Anticuerpos/sangre , Biomarcadores/sangre , Mapeo Cromosómico , Modelos Animales de Enfermedad , Predisposición Genética a la Enfermedad , Humanos , Hipertensión/diagnóstico , Hipertensión/genética , Hipertensión Renal/genética , Hipertensión Renal/inmunología , Hipertensión Renal/patología , Fragmentos Fc de Inmunoglobulinas/genética , Fragmentos Fc de Inmunoglobulinas/inmunología , Riñón/inmunología , Riñón/patología , Nefritis/genética , Nefritis/inmunología , Nefritis/patología , Pronóstico , Sitios de Carácter Cuantitativo , Ratas , Ratas Endogámicas SHR , Receptores Fc/genética , Receptores Fc/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA