Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Sci Rep ; 11(1): 24245, 2021 12 20.
Artigo em Inglês | MEDLINE | ID: mdl-34930978

RESUMO

Systemic lupus erythematosus (SLE) is a devastating autoimmune disorder characterized by failure of self-tolerance with resultant production of autoreactive antibodies. The etiology of this syndrome is complex, involving perturbations in immune cell signaling and development. The NZBWF1 mouse spontaneously develops a lupus-like syndrome and has been widely used as a model of SLE for over 60 years. The NZBWF1 model represents the F1 generation of a cross between New Zealand Black (NZB) and New Zealand White (NZW) mice. In order to better understand the factors that contribute to the development of autoimmunity, single cell RNA sequencing was conducted using the bone marrow from female NZBWF1 mice prior to the development of overt disease. The results were contrasted with single cell RNA sequencing results from the two parental strains. The expected findings of B cell abundance and upregulation, and evidence of interferon signaling were validated in this model. In addition, several novel areas of inquiry were identified. Most notably, the data showed a marked upregulation of the ferritin light chain across all cell types in the NZBWF1 mice compared to parental controls. This data can serve as a gene expression atlas of all hematopoietic cells in the NZBWF1 bone marrow prior to the development of autoimmunity.


Assuntos
Doenças Autoimunes/imunologia , Sequência de Bases , Lúpus Eritematoso Sistêmico/imunologia , Animais , Autoanticorpos/imunologia , Autoimunidade , Linfócitos B , Modelos Animais de Doenças , Feminino , Perfilação da Expressão Gênica , Tolerância Imunológica , Funções Verossimilhança , Camundongos , Fenótipo , RNA/análise
2.
Am J Physiol Renal Physiol ; 320(6): F1093-F1105, 2021 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-33843272

RESUMO

Hypertension and diabetes are the greatest factors influencing the progression of chronic kidney disease (CKD). Investigation into the role of nephron number in CKD alone or with hypertension has revealed a strong inverse relationship between the two; however, not much is known about the connection between nephron number and diabetic kidney disease. The heterogeneous stock-derived model of unilateral renal agenesis (HSRA) rat, a novel model of nephron deficiency, provides a unique opportunity to study the association between nephron number and hypertension and diabetes on CKD. HSRA rats exhibit failure of one kidney to develop in 50-75% of offspring, whereas the remaining offspring are born with two kidneys. Rats born with one kidney (HSRA-S) develop significant renal injury with age compared with two-kidney littermates (HSRA-C). The induction of hypertension as a secondary stressor leads to significantly more renal injury in HSRA-S compared with HSRA-C rats and nephrectomized HSRA-C (HSRA-UNX) rats. The present study sought to address the hypothesis that nephron deficiency in the HSRA rat would hasten renal injury in the presence of a secondary stressor of hyperglycemia. HSRA animals did not exhibit diabetes-related traits at any age; thus, streptozotocin (STZ) was used to induce hyperglycemia in HSRA-S, HSRA-C, and HSRA-UNX rats. STZ- and vehicle-treated animals were followed for 15 wk. STZ-treated animals developed robust hyperglycemia, but in contrast to the response to hypertension, neither HSRA-S nor HSRA-UNX animals developed proteinuria compared with vehicle treatment. In total, our data indicate that hyperglycemia from STZ alone does not have a significant impact on the onset or progression of injury in young one-kidney HSRA animals.NEW & NOTEWORTHY The HSRA rat, a novel model of nephron deficiency, provides a unique opportunity to study the association between nephron number and confounding cardiovascular complications that impact kidney health. Although hypertension was previously shown to exacerbate renal injury in young HSRA animals, diabetic hyperglycemia did not lead to worse renal injury, suggesting that nephron number has limited impact on kidney injury, at least in this model.


Assuntos
Envelhecimento , Diabetes Mellitus Experimental/complicações , Nefropatias Diabéticas/patologia , Rim Único/metabolismo , Animais , Hiperglicemia , Rim/fisiopatologia , Masculino , Ratos , Ratos Endogâmicos
3.
Physiol Genomics ; 53(3): 125-136, 2021 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-33491590

RESUMO

Preeclampsia is a progressive hypertensive disorder of pregnancy affecting 2%-8% of pregnancies globally. Preexisting chronic hypertension is a major risk factor associated with developing preeclampsia, and growing evidence suggests a role for the gut microbiome in the development of preeclampsia. However, neither alterations in the gut microbiome associated with preeclampsia nor the mechanisms involved are fully understood. In this study, we tested the hypothesis that normal gestational maternal gut microbiome remodeling is impaired in the Dahl salt-sensitive (Dahl S) rat model of superimposed preeclampsia. Gut microbiome profiles of pregnant Dahl S, normal pregnant Sprague-Dawley (SD), and matched virgin controls were assessed by 16S rRNA gene sequencing at baseline; during early, middle, and late pregnancy; and 1-wk postpartum. Dahl S rats had significantly higher abundance in Proteobacteria, and multiple genera were significantly different from SD rats at baseline. The pregnant SD displayed a significant increase in Proteobacteria and genera such as Helicobacter, but these were not different between pregnant and virgin Dahl S rats. By late pregnancy, Dahl S rats had significantly lower α-diversity and Firmicutes compared with their virgin Dahl S controls. ß-diversity was significantly different among groups (P < 0.001). KEGG metabolic pathways including those associated with short-chain fatty acids were different in Dahl S pregnancy but not in SD pregnancy. These results reveal an association between chronic hypertension and gut microbiome dysbiosis which may hinder pregnancy-specific remodeling in the gut microbial composition during superimposed preeclampsia.


Assuntos
Modelos Animais de Doenças , Microbioma Gastrointestinal/fisiologia , Hipertensão/fisiopatologia , Pré-Eclâmpsia/fisiopatologia , Animais , Bactérias/classificação , Bactérias/genética , Doença Crônica , Disbiose/genética , Disbiose/microbiologia , Disbiose/fisiopatologia , Feminino , Microbioma Gastrointestinal/genética , Variação Genética , Humanos , Filogenia , Gravidez , RNA Ribossômico 16S/genética , Ratos , Ratos Endogâmicos Dahl , Ratos Sprague-Dawley , Especificidade da Espécie
4.
Sci Rep ; 11(1): 2071, 2021 01 22.
Artigo em Inglês | MEDLINE | ID: mdl-33483609

RESUMO

Chronic kidney disease (CKD), which can ultimately progress to kidney failure, is influenced by genetics and the environment. Genes identified in human genome wide association studies (GWAS) explain only a small proportion of the heritable variation and lack functional validation, indicating the need for additional model systems. Outbred heterogeneous stock (HS) rats have been used for genetic fine-mapping of complex traits, but have not previously been used for CKD traits. We performed GWAS for urinary protein excretion (UPE) and CKD related serum biochemistries in 245 male HS rats. Quantitative trait loci (QTL) were identified using a linear mixed effect model that tested for association with imputed genotypes. Candidate genes were identified using bioinformatics tools and targeted RNAseq followed by testing in a novel in vitro model of human tubule, hypoxia-induced damage. We identified two QTL for UPE and five for serum biochemistries. Protein modeling identified a missense variant within Septin 8 (Sept8) as a candidate for UPE. Sept8/SEPTIN8 expression increased in HS rats with elevated UPE and tubulointerstitial injury and in the in vitro hypoxia model. SEPTIN8 is detected within proximal tubule cells in human kidney samples and localizes with acetyl-alpha tubulin in the culture system. After hypoxia, SEPTIN8 staining becomes diffuse and appears to relocalize with actin. These data suggest a role of SEPTIN8 in cellular organization and structure in response to environmental stress. This study demonstrates that integration of a rat genetic model with an environmentally induced tubule damage system identifies Sept8/SEPTIN8 and informs novel aspects of the complex gene by environmental interactions contributing to CKD risk.


Assuntos
Túbulos Renais/patologia , Rim/patologia , Septinas/genética , Animais , Hipóxia Celular , Efeito Fundador , Haplótipos , Humanos , Masculino , Ratos
5.
Physiol Rep ; 8(13): e14440, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32652814

RESUMO

The burden of hypertension in the United States is increasing and yields significant morbidity and mortality, and sex differences in hypertension are widely recognized. Reduced nitric oxide (NO) bioavailability and increased oxidative stress are known to contribute to the pathogenesis of hypertensive renal injury, and but their contributions to sex differences in injury progression of are undefined. Our purpose was to test the hypothesis that male hypertensive rats have accelerated renal injury compared to females and to determine the contributions of the nitric oxide pathway and oxidative stress in these differences. Male and female Dahl SS/Jr rats, a model that spontaneously develops hypertension with age, were allowed to age on a 0.3% NaCl diet until 3 or 6 months of age, at which points blood pressure was measured and plasma, tissue, and urine were collected. While no significant sex differences in blood pressure were present at either time point, renal injury measured by urine protein excretion was more severe (male = 44.9 ± 6; female = 15±3 mg/day/100 g bw, p = .0001), and renal function was reduced (male = 0.48 ± 0.02; female = 0.7 ± 0.03 ml min-1  g-1 kw, p = .001) in males compared to females with age. Both male and female rats exhibited reduced nitric oxide metabolites (3 months: male = 0.65 ± 0.1; female = 0.74 ± 0.3; 6 months: male = 0.16 ± 0.1; female = 0.41 ± 0.1 ml min-1  g-1 kw, p, age = 0.02, p, sex = 0.3). Levels of urinary TBARS were similar (3 months: male = 20±1.5; female = 23±1.8; 6 months: male = 26±4.8; female = 23±4.7µM day g-1 kw, p, age = 0.4, p, sex = 0.9), extracellular superoxide dismutase (EC SOD) mRNA was greater in females (3 months: male = 0.35 ± 0.03; female = 1.4 ± 0.2; 6 months: male = 0.4 ± 0.05; female = 1.3 ± 0.1 normalized counts, p, age = 0.7, p, sex < 0.0001), but EC SOD protein expression was not different (3 months: male = 0.01 ± 0.002; female = 0.01 ± 0.002; 6 months: male = 0.02 ± 0.004; female = 0.01 ± 0.002 relative density, p, age = 0.2, p, sex = 0.8). These data support the presence of significant sex differences in renal injury and function in the Dahl S rat and identify a need for further study into the mechanisms involved.


Assuntos
Hipertensão Renal/metabolismo , Óxido Nítrico/metabolismo , Estresse Oxidativo , Caracteres Sexuais , Animais , Feminino , Hipertensão Renal/fisiopatologia , Masculino , Ratos , Ratos Endogâmicos Dahl
6.
Hypertension ; 75(4): 1012-1024, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32148127

RESUMO

Arhgef11 is a Rho-guanine nucleotide exchange factor that was previously implicated in kidney injury in the Dahl salt-sensitive (SS) rat, a model of hypertension-related chronic kidney disease. Reduced Arhgef11 expression in an SS-Arhgef11SHR-minimal congenic strain (spontaneously hypertensive rat allele substituted for S allele) significantly decreased proteinuria, fibrosis, and improved renal hemodynamics, without impacting blood pressure compared with the control SS (SS-wild type). Here, SS-Arhgef11-/- and SS-wild type rats were placed on either low or elevated salt (0.3% or 2% NaCl) from 4 to 12 weeks of age. On low salt, starting at week 6 and through week 12, SS-Arhgef11-/- animals demonstrated a 3-fold decrease in proteinuria compared with SS-wild type. On high salt, beginning at week 6, SS-Arhgef11-/- animals demonstrated >2-fold lower proteinuria from weeks 8 to 12 and 30 mm Hg lower BP compared with SS-wild type. To better understand the molecular mechanisms of the renal protection from loss of Arhgef11, both RNA sequencing and discovery proteomics were performed on kidneys from week 4 (before onset of renal injury/proteinuria between groups) and at week 12 (low salt). The omics data sets revealed loss of Arhgef11 (SS-Arhgef11-/-) initiates early transcriptome/protein changes in the cytoskeleton starting as early as week 4 that impact a number of cellular functions, including actin cytoskeletal regulation, mitochondrial metabolism, and solute carrier transporters. In summary, in vivo phenotyping coupled with a multi-omics approach provides strong evidence that increased Arhgef11 expression in the Dahl SS rat leads to actin cytoskeleton-mediated changes in cell morphology and cell function that promote kidney injury, hypertension, and decline in kidney function.


Assuntos
Fatores de Troca do Nucleotídeo Guanina/genética , Hipertensão/genética , Rim/metabolismo , Proteinúria/genética , Insuficiência Renal Crônica/genética , Animais , Pressão Sanguínea/fisiologia , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Hipertensão/metabolismo , Masculino , Proteinúria/metabolismo , Ratos , Ratos Endogâmicos Dahl , Insuficiência Renal Crônica/metabolismo
7.
Am J Physiol Renal Physiol ; 318(4): F911-F921, 2020 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-32068459

RESUMO

The present study examined whether development of renal injury in the nondiabetic obese Dahl salt-sensitive leptin receptor mutant (SSLepRmutant) strain is associated with elevations in glomerular filtration rate and renal lipid accumulation. Baseline mean arterial pressure at 6 wk of age was similar between Dahl salt-sensitive wild-type (SSWT) and SSLepRmutant rats. However, by 18 wk of age, the SSLepRmutant strain developed hypertension, while the elevation in mean arterial pressure was not as severe in SSWT rats (192 ± 4 and 149 ± 6 mmHg, respectively). At baseline, proteinuria was fourfold higher in SSLepRmutant than SSWT rats and remained elevated throughout the study. The early development of progressive proteinuria was associated with renal hyperfiltration followed by a decline in renal function over the course of study in the SSLepRmutant compared with SSWT rats. Kidneys from the SSLepRmutant strain displayed more glomerulosclerosis and glomerular lipid accumulation than SSWT rats. Glomeruli were isolated from the renal cortex of both strains at 6 and 18 wk of age, and RNA sequencing was performed to identify genes and pathways driving glomerular injury. We observed significant increases in expression of the influx lipid transporters, chemokine (C-X-C motif) ligand 16 (Cxcl16) and scavenger receptor and fatty acid translocase (Cd36), respectively, and a significant decrease in expression of the efflux lipid transporter, ATP-binding cassette subfamily A member 2 (Abca2; cholesterol efflux regulatory protein 2), in SSLepRmutant compared with SSWT rats at 6 and 18 wk of age, which were validated by RT-PCR analysis. These data suggest an association between glomerular hyperfiltration and glomerular lipid accumulation during the early development of proteinuria associated with obesity.


Assuntos
Tecido Adiposo/metabolismo , Hemodinâmica , Hipertensão/metabolismo , Nefropatias/metabolismo , Glomérulos Renais/metabolismo , Metabolismo dos Lipídeos , Mutação , Obesidade/metabolismo , Receptores para Leptina/genética , Circulação Renal , Transportador 1 de Cassete de Ligação de ATP/genética , Transportador 1 de Cassete de Ligação de ATP/metabolismo , Transportadores de Cassetes de Ligação de ATP/genética , Transportadores de Cassetes de Ligação de ATP/metabolismo , Tecido Adiposo/patologia , Tecido Adiposo/fisiopatologia , Adiposidade , Animais , Antígenos CD36/genética , Antígenos CD36/metabolismo , Quimiocina CXCL16/genética , Quimiocina CXCL16/metabolismo , Modelos Animais de Doenças , Predisposição Genética para Doença , Taxa de Filtração Glomerular , Hipertensão/genética , Hipertensão/patologia , Hipertensão/fisiopatologia , Nefropatias/genética , Nefropatias/patologia , Nefropatias/fisiopatologia , Glomérulos Renais/patologia , Glomérulos Renais/fisiopatologia , Metabolismo dos Lipídeos/genética , Obesidade/genética , Obesidade/patologia , Obesidade/fisiopatologia , Fenótipo , Proteinúria/metabolismo , Proteinúria/patologia , Proteinúria/fisiopatologia , Ratos Endogâmicos Dahl , Cloreto de Sódio na Dieta
8.
Kidney360 ; 1(6): 510-520, 2020 06 25.
Artigo em Inglês | MEDLINE | ID: mdl-35368603

RESUMO

Background: Preeclampsia is a disorder of pregnancy with accompanying high disease and economic burdens in the United States. Evidence supporting longstanding effects of preeclampsia on the offspring of affected pregnancies is high, but the effects of current antihypertensive therapies for preeclampsia on cardio-renal outcomes are largely unknown. The purpose of this study was to test the hypothesis that sildenafil citrate, a phosphodiesterase-5 inhibitor, reprograms the risk of hypertension and kidney disease in offspring of preeclamptic pregnancies by altering responses to secondary stressors. Methods: Dahl SS/Jr rats on a 0.3% NaCl diet were mated. At gestational day 10, pregnant dams were randomized to vehicle diet or diet with sildenafil (50 mg/kg per day), which was continued until birth. Pups were weaned at 4 weeks of age and allowed to age on a 0.3% NaCl diet until 3 months of age. At this point, pups were randomized into three groups: baseline or no intervention, 2% NaCl diet challenge for 4 weeks, or a subpressor infusion of angiotensin II (200 ng/kg per minute) for 2 weeks. Results: There were no differences among maternal treatment groups at baseline. Upon introduction of 2% NaCl diet, male offspring of sildenafil-treated dams exhibited an attenuated rise in BP; however, this protection was not observed during angiotensin II infusion. Conclusions: Our findings indicate that intrapartum sildenafil does not reprogram the risk of hypertension and kidney disease in offspring of preeclamptic pregnancies.


Assuntos
Hipertensão , Pré-Eclâmpsia , Insuficiência Renal Crônica , Animais , Feminino , Hipertensão/tratamento farmacológico , Masculino , Gravidez , Ratos , Ratos Endogâmicos Dahl , Citrato de Sildenafila/farmacologia
9.
Physiol Genomics ; 52(1): 56-70, 2020 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-31841396

RESUMO

The HSRA rat is a model of congenital abnormalities of the kidney and urogenital tract (CAKUT). Our laboratory has used this model to investigate the role of nephron number (functional unit of the kidney) in susceptibility to develop kidney disease as 50-75% offspring are born with a single kidney (HSRA-S), while 25-50% are born with two kidneys (HSRA-C). HSRA-S rats develop increased kidney injury and hypertension with age compared with nephrectomized two-kidney animals (HSRA-UNX), suggesting that even slight differences in nephron number can be an important driver in decline in kidney function. The HSRA rat was selected and inbred from a family of outbred heterogeneous stock (NIH-HS) rats that exhibited a high incidence of CAKUT. The HS model was originally developed from eight inbred strains (ACI, BN, BUF, F344, M520, MR, WKY, and WN). The genetic make-up of the HSRA is therefore a mosaic of these eight inbred strains. Interestingly, the ACI progenitor of the HS model exhibits CAKUT in 10-15% of offspring with the genetic cause being attributed to the presence of a long-term repeat (LTR) within exon 1 of the c-Kit gene. Our hypothesis is that the HSRA and ACI share this common genetic cause, but other alleles in the HSRA genome contribute to the increased penetrance of CAKUT (75% HSRA vs. 15% in ACI). To facilitate genetic studies and better characterize the model, we sequenced the whole genome of the HSRA to a depth of ~50×. A genome-wide variant analysis of high-impact variants identified a number of novel genes that could be linked to CAKUT in the HSRA model. In summary, the identification of new genes/modifiers that lead to CAKUT/loss of one kidney in the HSRA model will provide greater insight into association between kidney development and susceptibility to develop cardiovascular disease later in life.


Assuntos
Estudos de Associação Genética , Predisposição Genética para Doença , Néfrons/embriologia , Organogênese/genética , Anormalidades Urogenitais/genética , Refluxo Vesicoureteral/genética , Sequenciamento Completo do Genoma , Animais , Sequência de Bases , Cromossomos de Mamíferos/genética , Modelos Animais de Doenças , Genoma , Genoma Mitocondrial , Íntrons/genética , Mitocôndrias/genética , Filogenia , Proteínas Proto-Oncogênicas c-kit/metabolismo , Ratos
10.
J Vis Exp ; (147)2019 05 22.
Artigo em Inglês | MEDLINE | ID: mdl-31180342

RESUMO

Nephron endowment refers to the total number of nephrons an individual is born with, as nephrogenesis in humans is completed by 36 weeks of gestation and no new nephrons are formed post-birth. Nephron number refers to the total number of nephrons measured at any point in time post-birth. Both genetic and environmental factors influence both nephron endowment and number. Understanding how specific genes or factors influence the process of nephrogenesis and nephron loss or demise is important as individuals with lower nephron endowment or number are thought to be at a higher risk of developing renal or cardiovascular disease. Understanding how environmental exposures over the course of a person's lifetime affects nephron number will also be vital in determining future disease risk. Thus, the ability to assess whole kidney nephron number quickly and reliably is a basic experimental requirement to better understand mechanisms that contribute to or promote nephrogenesis or nephron loss. Here, we describe the acid maceration method for the estimation of whole kidney nephron number based on the procedure described by Damadian, Shawayri, and Bricker, with slight modifications. The acid maceration method provides fast and reliable estimates of nephron number (as assessed by counting glomeruli) that are within 5% of those determined using more advanced, albeit expensive, methods such as magnetic resonance imaging. Moreover, the acid maceration method is an excellent high-throughput method to assess nephron number in large numbers of samples or experimental conditions.


Assuntos
Técnicas Citológicas/métodos , Rim/anatomia & histologia , Néfrons/citologia , Animais , Rim/citologia , Glomérulos Renais/citologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL
11.
Physiol Genomics ; 51(8): 342-355, 2019 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-31125289

RESUMO

Preeclampsia (PE), a multifactorial pregnancy-specific syndrome accounting for up to 8% of pregnancy complications, is a leading cause of maternal and fetal morbidity and mortality. PE is also associated with long-term risk of hypertension and stroke for both mother and fetus. Currently, the only "cure" is delivery of the baby and placenta, largely because the pathogenesis of PE is not yet fully understood. PE is associated with impaired vascular remodeling at the maternal-fetal interface and placental insufficiency; however, specific factors contributing to this impairment have not been identified. To identify molecular pathways involved in PE, we examined temporal transcriptomic changes occurring within the uterus, uterine implantation sites, and placentae from the Dahl salt-sensitive (Dahl S) rat model of superimposed PE compared with Sprague Dawley (SD) rats. We hypothesized that targeted gene analysis and whole transcriptome analysis would identify genetic factors that contribute to development of the preeclamptic phenotype in the Dahl S rat and unveil novel biomarkers, therapeutic targets, and mechanistic pathways in PE. Quantitative real-time PCR (qRT-PCR) and whole genome microarray analysis were performed on isolated total RNA from uterus (day 0), uterine implantation sites (days 7 and 10), and placenta (days 14 and 20). We found 624, 332, 185, and 366 genes to be differentially expressed between Dahl S (PE) and SD (normal pregnancy) on days 0, 7, 10, and 14, respectively. Our data revealed numerous pathways that may play a role in the pathophysiology of spontaneous superimposed PE and allow for further investigation of novel therapeutic targets and biomarker development.


Assuntos
Cronologia como Assunto , Perfilação da Expressão Gênica/métodos , Pré-Eclâmpsia/genética , Gravidez/genética , Transcriptoma , Animais , Sequência de Bases/genética , Biomarcadores , Modelos Animais de Doenças , Feminino , Placenta/metabolismo , Ratos , Ratos Endogâmicos Dahl , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase em Tempo Real , Transdução de Sinais/genética , Útero/metabolismo , Sequenciamento Completo do Genoma
12.
J Cell Sci ; 131(6)2018 03 22.
Artigo em Inglês | MEDLINE | ID: mdl-29507111

RESUMO

Reduced levels of the cardiac human (h)ERG ion channel protein and the corresponding repolarizing current IKr can cause arrhythmia and sudden cardiac death, but the underlying cellular mechanisms controlling hERG surface expression are not well understood. Here, we identified TRIOBP-1, an F-actin-binding protein previously associated with actin polymerization, as a putative hERG-interacting protein in a yeast-two hybrid screen of a cardiac library. We corroborated this interaction by performing Förster resonance energy transfer (FRET) in HEK293 cells and co-immunoprecipitation in HEK293 cells and native cardiac tissue. TRIOBP-1 overexpression reduced hERG surface expression and current density, whereas reducing TRIOBP-1 expression via shRNA knockdown resulted in increased hERG protein levels. Immunolabeling in rat cardiomyocytes showed that native TRIOBP-1 colocalized predominantly with myosin-binding protein C and secondarily with rat ERG. In human stem cell-derived cardiomyocytes, TRIOBP-1 overexpression caused intracellular co-sequestration of hERG signal, reduced native IKr and disrupted action potential repolarization. Ca2+ currents were also somewhat reduced and cell capacitance was increased. These findings establish that TRIOBP-1 interacts directly with hERG and can affect protein levels, IKr magnitude and cardiac membrane excitability.


Assuntos
Proteínas dos Microfilamentos/metabolismo , Miócitos Cardíacos/metabolismo , Animais , Cálcio/metabolismo , Células HEK293 , Humanos , Masculino , Proteínas dos Microfilamentos/genética , Ligação Proteica , Transporte Proteico , Ratos , Regulador Transcricional ERG/genética , Regulador Transcricional ERG/metabolismo
13.
Science ; 358(6368): 1332-1336, 2017 12 08.
Artigo em Inglês | MEDLINE | ID: mdl-29217578

RESUMO

Progressive kidney diseases are often associated with scarring of the kidney's filtration unit, a condition called focal segmental glomerulosclerosis (FSGS). This scarring is due to loss of podocytes, cells critical for glomerular filtration, and leads to proteinuria and kidney failure. Inherited forms of FSGS are caused by Rac1-activating mutations, and Rac1 induces TRPC5 ion channel activity and cytoskeletal remodeling in podocytes. Whether TRPC5 activity mediates FSGS onset and progression is unknown. We identified a small molecule, AC1903, that specifically blocks TRPC5 channel activity in glomeruli of proteinuric rats. Chronic administration of AC1903 suppressed severe proteinuria and prevented podocyte loss in a transgenic rat model of FSGS. AC1903 also provided therapeutic benefit in a rat model of hypertensive proteinuric kidney disease. These data indicate that TRPC5 activity drives disease and that TRPC5 inhibitors may be valuable for the treatment of progressive kidney diseases.


Assuntos
Glomerulosclerose Segmentar e Focal/tratamento farmacológico , Hipertensão Renal/tratamento farmacológico , Indazóis/farmacologia , Proteinúria/tratamento farmacológico , Canais de Cátion TRPC/antagonistas & inibidores , Animais , Modelos Animais de Doenças , Glomerulosclerose Segmentar e Focal/genética , Mutação , Podócitos/efeitos dos fármacos , Ratos , Ratos Endogâmicos Dahl , Ratos Transgênicos , Bibliotecas de Moléculas Pequenas , Canais de Cátion TRPC/farmacologia , Proteínas rac1 de Ligação ao GTP/genética
14.
Am J Physiol Renal Physiol ; 311(4): F793-F804, 2016 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-27465994

RESUMO

The current study examined the effect of obesity on the development of renal injury within the genetic background of the Dahl salt-sensitive rat with a dysfunctional leptin receptor derived from zinc-finger nucleases (SSLepRmutant strain). At 6 wk of age, body weight was 35% higher in the SSLepRmutant strain compared with SSWT rats and remained elevated throughout the entire study. The SSLepRmutant strain exhibited impaired glucose tolerance and increased plasma insulin levels at 6 wk of age, suggesting insulin resistance while SSWT rats did not. However, blood glucose levels were normal throughout the course of the study. Systolic arterial pressure (SAP) was similar between the two strains from 6 to 10 wk of age. However, by 18 wk of age, the development of hypertension was more severe in the SSLepRmutant strain compared with SSWT rats (201 ± 10 vs. 155 ± 3 mmHg, respectively). Interestingly, proteinuria was substantially higher at 6 wk of age in the SSLepRmutant strain vs. SSWT rats (241 ± 27 vs. 24 ± 2 mg/day, respectively) and remained elevated until the end of the study. The kidneys from the SSLepRmutant strain displayed significant glomerular injury, including podocyte foot process effacement and lipid droplets compared with SSWT rats as early as 6 wk of age. By 18 wk of age, plasma creatinine levels were twofold higher in the SSLepRmutant strain vs. SSWT rats, suggesting the presence of chronic kidney disease (CKD). Overall, these results indicate that the SSLepRmutant strain develops podocyte injury and proteinuria independently of hyperglycemia and elevated arterial pressure that later progresses to CKD.


Assuntos
Pressão Arterial/fisiologia , Hiperglicemia/patologia , Obesidade/patologia , Podócitos/patologia , Receptores para Leptina/genética , Insuficiência Renal Crônica/patologia , Animais , Glicemia/metabolismo , Hiperglicemia/genética , Hiperglicemia/metabolismo , Rim/metabolismo , Rim/patologia , Masculino , Obesidade/genética , Obesidade/metabolismo , Podócitos/metabolismo , Ratos , Ratos Endogâmicos Dahl , Ratos Transgênicos , Receptores para Leptina/metabolismo , Insuficiência Renal Crônica/genética , Insuficiência Renal Crônica/metabolismo
15.
Am J Physiol Renal Physiol ; 310(10): F1054-64, 2016 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-26936874

RESUMO

There is little clinical data of how hypertension may influence individuals with nephron deficiency in the context of being born with a single kidney. We recently developed a new rat model (the heterogeneous stock-derived model of unilateral renal agenesis rat) that is born with a single kidney and exhibits progressive kidney injury and decline in kidney function with age. We hypothesized that DOCA-salt would induce a greater increase in blood pressure and therefore accelerate the progression of kidney injury in rats born with a solitary kidney compared with rats that have undergone unilateral nephrectomy. Time course evaluation of blood pressure, kidney injury, and renal hemodynamics was performed in the following six groups of animals from weeks 13 to 18: 1) DOCA-treated rats with a solitary kidney (DOCA+S group), 2) placebo-treated rats with a solitary kidney, 3) DOCA-treated control rats with two kidneys (DOCA+C group), 4) placebo-treated control rats with two kidneys, 5) DOCA-treated rats with two kidneys that underwent uninephrectomy (DOCA+UNX8 group), and 6) placebo-treated rats with two kidneys that underwent uninephrectomy. DOCA+S rats demonstrated a significant rise (P < 0.05) in blood pressure (192 ± 4 mmHg), proteinuria (205 ± 31 mg/24 h), and a decline in glomerular filtration rate (600 ± 42 µl·min(-1)·g kidney weight(-1)) relative to the DOCA+UNX8 (173 ± 3 mmHg, 76 ± 26 mg/24 h, and 963 ± 36 µl·min(-1)·g kidney weight(-1)) and DOCA+C (154 ± 2 mmHg, 7 ± 1 mg/24 h, and 1,484 ± 121 µl·min(-1)·g kidney weight(-1)) groups. Placebo-treated groups showed no significant change among the three groups. An assessment of renal injury markers via real-time PCR/Western blot analysis and histological analysis was concordant with the measured physiological parameters. In summary, congenital solitary kidney rats are highly susceptible to the induction of hypertension compared with uninephrectomized rats, suggesting that low nephron endowment is an important driver of elevated blood pressure, hastening nephron injury through the transmission of elevated systemic blood pressure and thereby accelerating decline in kidney function.


Assuntos
Hipertensão/induzido quimicamente , Nefropatias/congênito , Rim/anormalidades , Insuficiência Renal/etiologia , Animais , Anormalidades Congênitas , Acetato de Desoxicorticosterona , Progressão da Doença , Feminino , Hipertensão/complicações , Hipertensão/patologia , Rim/patologia , Nefropatias/complicações , Masculino , Miocárdio/patologia , Nefrectomia , Ratos , Sódio
16.
PLoS One ; 10(7): e0132553, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26172442

RESUMO

Previously, genetic analyses identified that variants in Arhgef11 may influence kidney injury in the Dahl salt-sensitive (S) rat, a model of hypertensive chronic kidney disease. To understand the potential mechanism by which altered expression and/or protein differences in Arhgef11 could play a role in kidney injury, stably transduced Arhgef11 knockdown cell lines as well as primary cultures of proximal tubule cells were studied. Genetic knockdown of Arhgef11 in HEK293 and NRK resulted in reduced RhoA activity, decreased activation of Rho-ROCK pathway, and less stress fiber formation versus control, similar to what was observed by pharmacological inhibition (fasudil). Primary proximal tubule cells (PTC) cultured from the S exhibited increased expression of Arhgef11, increased RhoA activity, and up regulation of Rho-ROCK signaling compared to control (small congenic). The cells were also more prone (versus control) to TGFß-1 induced epithelial-mesenchymal transition (EMT), a hallmark feature of the development of renal interstitial fibrosis, and characterized by development of spindle shape morphology, gene expression changes in EMT markers (Col1a3, Mmp9, Bmp7, and Ocln) and increased expression of N-Cadherin and Vimentin. S derived PTC demonstrated a decreased ability to uptake FITC-albumin compared to the small congenic in vitro, which was confirmed by assessment of albumin re-uptake in vivo by infusion of FITC-albumin and immunofluorescence imaging. In summary, these studies suggest that genetic variants in the S form of Arhgef11 via increased expression and/or protein activity play a role in promoting kidney injury in the S rat through changes in cell morphology (Rho-Rock and/or EMT) that impact the function of tubule cells.


Assuntos
Fatores de Troca do Nucleotídeo Guanina/genética , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Insuficiência Renal Crônica/genética , Alelos , Animais , Animais Congênicos , Linhagem Celular , Modelos Animais de Doenças , Transição Epitelial-Mesenquimal/genética , Transição Epitelial-Mesenquimal/fisiologia , Técnicas de Silenciamento de Genes , Variação Genética , Fatores de Troca do Nucleotídeo Guanina/antagonistas & inibidores , Células HEK293 , Humanos , Túbulos Renais Proximais/metabolismo , Túbulos Renais Proximais/patologia , Masculino , Plaquinas/metabolismo , Ratos , Ratos Endogâmicos Dahl , Ratos Endogâmicos SHR , Insuficiência Renal Crônica/metabolismo , Insuficiência Renal Crônica/patologia , Fatores de Troca de Nucleotídeo Guanina Rho/antagonistas & inibidores , Fatores de Troca de Nucleotídeo Guanina Rho/genética , Fatores de Troca de Nucleotídeo Guanina Rho/metabolismo , Transdução de Sinais , Proteínas rho de Ligação ao GTP/metabolismo , Quinases Associadas a rho/metabolismo
17.
J Am Soc Nephrol ; 26(7): 1634-46, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25349207

RESUMO

Some studies have reported up to 40% of patients born with a single kidney develop hypertension, proteinuria, and in some cases renal failure. The increased susceptibility to renal injury may be due, in part, to reduced nephron numbers. Notably, children who undergo nephrectomy or adults who serve as kidney donors exhibit little difference in renal function compared with persons who have two kidneys. However, the difference in risk between being born with a single kidney versus being born with two kidneys and then undergoing nephrectomy are unclear. Animal models used previously to investigate this question are not ideal because they require invasive methods to model congenital solitary kidney. In this study, we describe a new genetic animal model, the heterogeneous stock-derived model of unilateral renal agenesis (HSRA) rat, which demonstrates 50%-75% spontaneous incidence of a single kidney. The HSRA model is characterized by reduced nephron number (more than would be expected by loss of one kidney), early kidney/glomerular hypertrophy, and progressive renal injury, which culminates in reduced renal function. Long-term studies of temporal relationships among BP, renal hemodynamics, and renal function demonstrate that spontaneous single-kidney HSRA rats are more likely than uninephrectomized normal littermates to exhibit renal impairment because of the combination of reduced nephron numbers and prolonged exposure to renal compensatory mechanisms (i.e., hyperfiltration). Future studies with this novel animal model may provide additional insight into the genetic contributions to kidney development and agenesis and the factors influencing susceptibility to renal injury in individuals with congenital solitary kidney.


Assuntos
Rim/anormalidades , Néfrons/fisiopatologia , Insuficiência Renal Crônica/etiologia , Anormalidades Urogenitais/complicações , Anormalidades Urogenitais/genética , Injúria Renal Aguda/etiologia , Injúria Renal Aguda/fisiopatologia , Adulto , Análise de Variância , Animais , Causalidade , Criança , Modelos Animais de Doenças , Progressão da Doença , Feminino , Taxa de Filtração Glomerular , Humanos , Hipertrofia/patologia , Masculino , Modelos Genéticos , Nefrectomia/efeitos adversos , Proteinúria/fisiopatologia , Distribuição Aleatória , Ratos , Insuficiência Renal Crônica/fisiopatologia , Medição de Risco
18.
J Am Soc Nephrol ; 25(11): 2499-510, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24722447

RESUMO

Nuclear hormone receptors of the NR4A subgroup have been implicated in cancer, atherosclerosis, and metabolic disease. However, little is known about the role of these receptors in kidney health or disease. Nr4a1-deficient rats (Nr4a1(-/-)) developed on a genetic background susceptible to kidney injury (fawn-hooded hypertensive rat [FHH]) were evaluated for BP, proteinuria, renal function, and metabolic parameters from 4 to 24 weeks-of-age. By week 24, Nr4a1(-/-) rats exhibited significantly higher proteinuria (approximately 4-fold) and decreased GFR compared with FHH controls. The severity of tubular atrophy, tubular casts, and interstitial fibrosis increased significantly in Nr4a1(-/-) rats and was accompanied by a large increase in immune cell infiltration, predominantly macrophages and to a lesser extent T cells and B cells. Global transcriptome and network analyses at weeks 8, 16, and 24 identified several proinflammatory genes and pathways differentially regulated between strains. Bone marrow crosstransplantation studies demonstrated that kidney injury in Nr4a1(-/-) rats was almost completely rescued by bone marrow transplanted from FHH controls. In vitro, macrophages isolated from Nr4a1(-/-) rats demonstrated increased immune activation compared with FHH-derived macrophages. In summary, the loss of Nr4a1 in immune cells appears to cause the increased kidney injury and reduced renal function observed in the Nr4a1(-/-) model.


Assuntos
Macrófagos/imunologia , Membro 1 do Grupo A da Subfamília 4 de Receptores Nucleares/genética , Membro 1 do Grupo A da Subfamília 4 de Receptores Nucleares/imunologia , Insuficiência Renal Crônica/genética , Insuficiência Renal Crônica/imunologia , Animais , Transplante de Medula Óssea , Células Cultivadas , Modelos Animais de Doenças , Ligação Genética , Predisposição Genética para Doença , Glomérulos Renais/imunologia , Glomérulos Renais/metabolismo , Túbulos Renais/imunologia , Túbulos Renais/metabolismo , Macrófagos/citologia , Masculino , Proteinúria/genética , Proteinúria/imunologia , Proteinúria/metabolismo , Ratos Endogâmicos , Ratos Mutantes , Insuficiência Renal Crônica/metabolismo , Transcriptoma
19.
Mamm Genome ; 24(3-4): 95-104, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23404175

RESUMO

Cataracts are a major cause of blindness. The most common forms of cataracts are age- and UV-related and develop mostly in the elderly, while congenital cataracts appear at birth or in early childhood. The Dahl salt-sensitive (SS/Jr) rat is an extensively used model of salt-sensitive hypertension that exhibits concomitant renal disease. In the mid-1980s, cataracts appeared in a few animals in the Dahl S colony, presumably the result of a spontaneous mutation. The mutation was fixed and bred to establish the SS/Jr-Ctr substrain. The SS/Jr-Ctr substrain has been used exclusively by a single investigator to study the role of steroids and hypertension. Using a classical positional cloning approach, we localized the cataract gene with high resolution to a less than 1-Mbp region on chromosome 9 using an F1(SS/Jr-Ctr × SHR) × SHR backcross population. The 1-Mbp region contained only 13 genes, including 4 genes from the γ-crystallins (Cryg) gene family, which are known to play a role in cataract formation. All of the γ-crystallins were sequenced and a novel point mutation in the start codon (ATG → GTG) of the Crygd gene was identified. This led to the complete absence of the CRYGD protein in the eyes of the SS/Jr-Ctr strain. In summary, the identification of the genetic cause in this novel cataract model may provide an opportunity to better understand the development of cataracts, particularly in the context of hypertension.


Assuntos
Catarata/genética , Códon de Iniciação/genética , Mutação , gama-Cristalinas/genética , Animais , Catarata/diagnóstico , Catarata/patologia , Clonagem Molecular , Feminino , Ligação Genética , Genótipo , Hipertensão/genética , Masculino , Locos de Características Quantitativas , Ratos , Ratos Endogâmicos SHR , Análise de Sequência de DNA
20.
Hypertension ; 60(5): 1157-68, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22987919

RESUMO

A previous genetic analysis comparing the Dahl salt-sensitive (S) rat with the spontaneously hypertensive rat identified a major locus on chromosome 2 that influences proteinuria in the S rat. In the present study, blood pressure, proteinuria, and renal hemodynamics were evaluated in congenic strains with small segments of the protective spontaneously hypertensive rat genome on the S background. Proteinuria and renal function were significantly improved in the congenic strains compared with the S. The causative locus interval was narrowed to <375 kb on the basis of congenic strains, haplotype data, comparative mapping, and concordance with human genetic studies. Sequencing of the coding region of genes in this region identified 36 single nucleotide polymorphisms (13 nonsynonymous and 23 synonymous). Gene expression profiling indicated that only a few genes exhibited differential expression. Arhgef11, Pear1, and Sh2d2 were identified as important candidate genes that may be linked to kidney injury in the S rat. In particular, Arhgef11 plays an important role in the activation of the Rho-ROCK signaling pathway. Inhibition of this pathway using fasudil resulted in a significant reduction of proteinuria in treated S rats (compared with untreated S). However, no difference was observed between treated or untreated spontaneously hypertensive rat or congenic strains. The homologous region in humans was found to be associated with estimated glomerular filtration rate in the Candidate Gene Association Resource population. In summary, these findings demonstrate that allelic variants in Arhgef11, acting through the Rho-ROCK pathway, could influence kidney injury in the S as well as provide insight into human kidney disease.


Assuntos
Predisposição Genética para Doença/genética , Fatores de Troca do Nucleotídeo Guanina/genética , Nefropatias/genética , Polimorfismo de Nucleotídeo Único , Ratos Endogâmicos Dahl/genética , 1-(5-Isoquinolinasulfonil)-2-Metilpiperazina/análogos & derivados , 1-(5-Isoquinolinasulfonil)-2-Metilpiperazina/farmacologia , Animais , Animais Congênicos , Pressão Sanguínea/genética , Western Blotting , Mapeamento Cromossômico , Perfilação da Expressão Gênica , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Humanos , Rim/irrigação sanguínea , Rim/metabolismo , Rim/fisiopatologia , Nefropatias/metabolismo , Nefropatias/fisiopatologia , Masculino , Inibidores de Proteínas Quinases/farmacologia , Proteinúria/genética , Proteinúria/metabolismo , Locos de Características Quantitativas/genética , Ratos , Ratos Endogâmicos Dahl/metabolismo , Ratos Endogâmicos SHR , Circulação Renal , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Quinases Associadas a rho/genética , Quinases Associadas a rho/metabolismo , Proteína rhoA de Ligação ao GTP/genética , Proteína rhoA de Ligação ao GTP/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA