Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Am J Physiol Renal Physiol ; 324(1): F30-F42, 2023 01 01.
Article in English | MEDLINE | ID: mdl-36264884

ABSTRACT

Collectrin (Tmem27), an angiotensin-converting enzyme 2 homologue, is a chaperone of amino acid transporters in the kidney and endothelium. Global collectrin knockout (KO) mice have hypertension, endothelial dysfunction, exaggerated salt sensitivity, and diminished renal blood flow. This phenotype is associated with altered nitric oxide and superoxide balance and increased proximal tubule (PT) Na+/H+ exchanger isoform 3 (NHE3) expression. Collectrin is located on the X chromosome where genome-wide association population studies have largely been excluded. In the present study, we generated PT-specific collectrin KO (PT KO) mice to determine the precise contribution of PT collectrin in blood pressure homeostasis. We also examined the association of human TMEM27 single-nucleotide polymorphisms with blood pressure traits in 11,926 Hispanic Community Health Study/Study of Latinos (HCHS/SOL) Hispanic/Latino participants. PT KO mice exhibited hypertension, and this was associated with increased baseline NHE3 expression and diminished lithium excretion. However, PT KO mice did not display exaggerated salt sensitivity or a reduction in renal blood flow compared with control mice. Furthermore, PT KO mice exhibited enhanced endothelium-mediated dilation, suggesting a compensatory response to systemic hypertension induced by deficiency of collectrin in the PT. In HCHS/SOL participants, we observed sex-specific single-nucleotide polymorphism associations with diastolic blood pressure. In conclusion, loss of collectrin in the PT is sufficient to induce hypertension, at least in part, through activation of NHE3. Importantly, our model supports the notion that altered renal blood flow may be a determining factor for salt sensitivity. Further studies are needed to investigate the role of the TMEM27 locus on blood pressure and salt sensitivity in humans.NEW & NOTEWORTHY The findings of our study are significant in several ways: 1) loss of an amino acid chaperone in the proximal tubule is sufficient to cause hypertension, 2) the results in global and proximal tubule-specific collectrin knockout mice support the notion that vascular dysfunction is required for salt sensitivity or that impaired renal tubule function causes hypertension but is not sufficient to cause salt sensitivity, and 3) our study is the first to implicate a role of collectrin in human hypertension.


Subject(s)
Blood Pressure , Hypertension , Kidney Tubules, Proximal , Membrane Glycoproteins , Animals , Female , Humans , Male , Mice , Blood Pressure/physiology , Genome-Wide Association Study , Hispanic or Latino/genetics , Hypertension/genetics , Kidney Tubules, Proximal/metabolism , Mice, Knockout , Sodium Chloride, Dietary/metabolism , Sodium-Hydrogen Exchanger 3/genetics , Sodium-Hydrogen Exchanger 3/metabolism , Membrane Glycoproteins/deficiency , Membrane Glycoproteins/genetics
3.
J Am Soc Nephrol ; 31(1): 102-116, 2020 01.
Article in English | MEDLINE | ID: mdl-31727850

ABSTRACT

BACKGROUND: GSTM1 encodes glutathione S-transferase µ-1 (GSTM1), which belongs to a superfamily of phase 2 antioxidant enzymes. The highly prevalent GSTM1 deletion variant is associated with kidney disease progression in human cohorts: the African American Study of Kidney Disease and Hypertension and the Atherosclerosis Risk in Communities (ARIC) Study. METHODS: We generated a Gstm1 knockout mouse line to study its role in a CKD model (involving subtotal nephrectomy) and a hypertension model (induced by angiotensin II). We examined the effect of intake of cruciferous vegetables and GSTM1 genotypes on kidney disease in mice as well as in human ARIC study participants. We also examined the importance of superoxide in the mediating pathways and of hematopoietic GSTM1 on renal inflammation. RESULTS: Gstm1 knockout mice displayed increased oxidative stress, kidney injury, and inflammation in both models. The central mechanism for kidney injury is likely mediated by oxidative stress, because treatment with Tempol, an superoxide dismutase mimetic, rescued kidney injury in knockout mice without lowering BP. Bone marrow crosstransplantation revealed that Gstm1 deletion in the parenchyma, and not in bone marrow-derived cells, drives renal inflammation. Furthermore, supplementation with cruciferous broccoli powder rich in the precursor to antioxidant-activating sulforaphane significantly ameliorated kidney injury in Gstm1 knockout, but not wild-type mice. Similarly, among humans (ARIC study participants), high consumption of cruciferous vegetables was associated with fewer kidney failure events compared with low consumption, but this association was observed primarily in participants homozygous for the GSTM1 deletion variant. CONCLUSIONS: Our data support a role for the GSTM1 enzyme in the modulation of oxidative stress, inflammation, and protective metabolites in CKD.


Subject(s)
Brassicaceae , Diet , Gene Deletion , Glutathione Transferase/genetics , Renal Insufficiency, Chronic/genetics , Vegetables , Animals , Disease Models, Animal , Female , Glutathione Transferase/physiology , Humans , Male , Mice , Middle Aged , Renal Insufficiency, Chronic/prevention & control
4.
Am J Physiol Renal Physiol ; 319(4): F654-F663, 2020 10 01.
Article in English | MEDLINE | ID: mdl-32715759

ABSTRACT

Acute kidney injury (AKI) is a frequent complication of sepsis and an important cause of morbidity and mortality worldwide. A cornerstone of sepsis-associated AKI is dysregulated inflammation, leading to increased tissue oxidative stress and free radical formation, which leads to multiple forms of cell death. DJ-1 is a peroxiredoxin protein with multiple functions, including its ability to control cellular oxidative stress. Although DJ-1 is expressed prominently by renal tubules, its role in AKI has not been investigated. In the present study, we examined the effect of DJ-1 deficiency in a murine model of endotoxin-induced AKI. Endotoxemia induced greater kidney injury in DJ-1-deficient mice. Furthermore, DJ-1 deficiency increased renal oxidative stress associated with increased renal tubular apoptosis and with expression of death domain-associated protein (DAXX). Similar to the in vivo model, in vitro experiments using a medullary collecting duct cell line (mIMCD3) and cytotoxic serum showed that serum obtained from wild-type mice resulted in increased expression of s100A8/s100A9, DAXX, and apoptosis in DJ-1-deficient mIMCD3 cells. Our findings demonstrate a novel renal protective role for renal tubular DJ-1 during endotoxemia through control of oxidative stress, renal inflammation, and DAXX-dependent apoptosis.


Subject(s)
Acute Kidney Injury/prevention & control , Endotoxemia/complications , Kidney Tubules/enzymology , Lipopolysaccharides , Nephritis/prevention & control , Protein Deglycase DJ-1/metabolism , Acute Kidney Injury/enzymology , Acute Kidney Injury/etiology , Acute Kidney Injury/pathology , Animals , Apoptosis , Calgranulin A/metabolism , Calgranulin B/metabolism , Cell Line , Co-Repressor Proteins/metabolism , Cytokines/metabolism , Disease Models, Animal , Kidney Tubules/pathology , Male , Mice, Inbred C57BL , Mice, Transgenic , Molecular Chaperones/metabolism , Nephritis/enzymology , Nephritis/etiology , Nephritis/pathology , Nitrosative Stress , Oxidative Stress , Protein Deglycase DJ-1/genetics , Signal Transduction
5.
Kidney Int ; 98(1): 100-115, 2020 07.
Article in English | MEDLINE | ID: mdl-32444136

ABSTRACT

Lupus nephritis is the end organ manifestation of systemic lupus erythematosus. Iron metabolism and its master regulator, hepcidin, are known to regulate cell proliferation and inflammation, but their direct role in the pathophysiology of lupus nephritis remains under-investigated. Exogenous hepcidin reduced the severity of lupus nephritis in MRL/lpr mice, a preclinical model of spontaneous systemic lupus erythematosus without worsening anemia of inflammation. Hepcidin treatment reduced renal iron accumulation, systemic and intrarenal cytokines, and renal immune cell infiltration, independent of glomerular immune complex deposits and circulating autoantibodies. Hepcidin increased renal H-ferritin (a ferroxidase), reduced expression of free iron dependent DNA synthesis enzymes, Ribonucleotide Reductase 1 and 2, and intra-renal macrophage proliferation. These findings were recapitulated in vitro upon treatment of macrophages with hepcidin and murine colony stimulation factor-1. Furthermore, hepcidin-treated macrophages secreted less IL-1ß and IL-6 upon stimulation with the TLR3 agonist polyinosine-polycytidylic acid. Of clinical relevance, hepcidin reduced progression and severity of nephritis in old mice with established systemic autoimmunity and overt proteinuria, highlighting its therapeutic potential. Thus, our findings provide a proof-of-concept that targeting cellular iron metabolism with hepcidin represents a promising therapeutic strategy in lupus nephritis.


Subject(s)
Lupus Erythematosus, Systemic , Lupus Nephritis , Animals , Disease Models, Animal , Hepcidins , Homeostasis , Iron , Lupus Nephritis/drug therapy , Mice , Mice, Inbred MRL lpr
6.
PLoS Genet ; 13(5): e1006728, 2017 May.
Article in English | MEDLINE | ID: mdl-28498854

ABSTRACT

Hypertension is a leading cause of global disease, mortality, and disability. While individuals of African descent suffer a disproportionate burden of hypertension and its complications, they have been underrepresented in genetic studies. To identify novel susceptibility loci for blood pressure and hypertension in people of African ancestry, we performed both single and multiple-trait genome-wide association analyses. We analyzed 21 genome-wide association studies comprised of 31,968 individuals of African ancestry, and validated our results with additional 54,395 individuals from multi-ethnic studies. These analyses identified nine loci with eleven independent variants which reached genome-wide significance (P < 1.25×10-8) for either systolic and diastolic blood pressure, hypertension, or for combined traits. Single-trait analyses identified two loci (TARID/TCF21 and LLPH/TMBIM4) and multiple-trait analyses identified one novel locus (FRMD3) for blood pressure. At these three loci, as well as at GRP20/CDH17, associated variants had alleles common only in African-ancestry populations. Functional annotation showed enrichment for genes expressed in immune and kidney cells, as well as in heart and vascular cells/tissues. Experiments driven by these findings and using angiotensin-II induced hypertension in mice showed altered kidney mRNA expression of six genes, suggesting their potential role in hypertension. Our study provides new evidence for genes related to hypertension susceptibility, and the need to study African-ancestry populations in order to identify biologic factors contributing to hypertension.


Subject(s)
Blood Pressure/genetics , Genetic Loci , Hypertension/genetics , Multifactorial Inheritance , Black or African American/genetics , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Cadherins/genetics , Case-Control Studies , Female , Genome-Wide Association Study , Humans , Hypertension/ethnology , Male , Membrane Proteins/genetics , Mice , Polymorphism, Single Nucleotide
7.
Am J Physiol Renal Physiol ; 316(6): F1141-F1150, 2019 06 01.
Article in English | MEDLINE | ID: mdl-30943068

ABSTRACT

Hypertension affects over 1 billion people worldwide and increases the risk for heart failure, stroke, and chronic kidney disease. Despite high prevalence and devastating impact, its etiology still remains poorly understood for most hypertensive cases. Rcn2, which encodes reticulocalbin 2, is a candidate gene for atherosclerosis that we have previously reported in mice. Here, we identified Rcn2 as a novel regulator of blood pressure in mice. Rcn2 was abundantly expressed in the endothelium and adventitia of normal arteries and was dramatically upregulated in the medial layer of the artery undergoing structural remodeling. Deletion of Rcn2 lowered basal blood pressure and attenuated ANG II-induced hypertension in C57BL/6 mice. siRNA knockdown of Rcn2 dramatically increased production of the nitric oxide (NO) breakdown products nitrite and nitrate by endothelial cells but not by smooth muscle cells. Isolated carotid arteries from Rcn2-/- mice showed an increased sensitivity to the ACh-induced NO-mediated relaxant response compared with arteries of Rcn2+/+ mice. Analysis of a recent meta-data set showed associations of genetic variants near RCN2 with blood pressure in humans. These data suggest that Rcn2 regulates blood pressure and contributes to hypertension through actions on endothelial NO synthase.


Subject(s)
Angiotensin II , Blood Pressure , Calcium-Binding Proteins/deficiency , Hypertension/prevention & control , Animals , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Cells, Cultured , Disease Models, Animal , Endothelial Cells/metabolism , Humans , Hypertension/chemically induced , Hypertension/metabolism , Hypertension/physiopathology , Kidney/metabolism , Kidney/physiopathology , Male , Mice, Inbred C57BL , Mice, Knockout , Nitric Oxide/metabolism , Nitric Oxide Synthase Type III/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction , Vasodilation
8.
Am J Hum Genet ; 99(3): 636-646, 2016 Sep 01.
Article in English | MEDLINE | ID: mdl-27588450

ABSTRACT

We analyzed genome-wide association studies (GWASs), including data from 71,638 individuals from four ancestries, for estimated glomerular filtration rate (eGFR), a measure of kidney function used to define chronic kidney disease (CKD). We identified 20 loci attaining genome-wide-significant evidence of association (p < 5 × 10(-8)) with kidney function and highlighted that allelic effects on eGFR at lead SNPs are homogeneous across ancestries. We leveraged differences in the pattern of linkage disequilibrium between diverse populations to fine-map the 20 loci through construction of "credible sets" of variants driving eGFR association signals. Credible variants at the 20 eGFR loci were enriched for DNase I hypersensitivity sites (DHSs) in human kidney cells. DHS credible variants were expression quantitative trait loci for NFATC1 and RGS14 (at the SLC34A1 locus) in multiple tissues. Loss-of-function mutations in ancestral orthologs of both genes in Drosophila melanogaster were associated with altered sensitivity to salt stress. Renal mRNA expression of Nfatc1 and Rgs14 in a salt-sensitive mouse model was also reduced after exposure to a high-salt diet or induced CKD. Our study (1) demonstrates the utility of trans-ethnic fine mapping through integration of GWASs involving diverse populations with genomic annotation from relevant tissues to define molecular mechanisms by which association signals exert their effect and (2) suggests that salt sensitivity might be an important marker for biological processes that affect kidney function and CKD in humans.


Subject(s)
Ethnicity/genetics , Genome-Wide Association Study , Kidney/physiopathology , Renal Insufficiency, Chronic/genetics , Renal Insufficiency, Chronic/physiopathology , Sodium Chloride/pharmacology , Stress, Physiological/drug effects , Stress, Physiological/genetics , Alleles , Animals , Deoxyribonuclease I/metabolism , Diabetes Mellitus/genetics , Disease Models, Animal , Drosophila melanogaster/genetics , Female , Glomerular Filtration Rate/genetics , Humans , Kidney/pathology , Linkage Disequilibrium , Male , NFATC Transcription Factors/genetics , Polymorphism, Single Nucleotide/genetics , Quantitative Trait Loci , RGS Proteins/genetics , Racial Groups/genetics , Salt Tolerance/genetics , Sodium-Phosphate Cotransporter Proteins, Type IIa/genetics
9.
J Immunol ; 198(6): 2341-2351, 2017 03 15.
Article in English | MEDLINE | ID: mdl-28159899

ABSTRACT

Cross-presentation is a modular series of intracellular events dictating the internalization and subsequent MHC class I (MHC I) display of extracellular Ags. This process has been defined in dendritic cells and plays a fundamental role in the induction of CD8+ T cell immunity during viral, intracellular bacterial, and antitumor responses. Herein, acute viral infection of murine liver with adenovirus, a model for intrahepatic cross-presentation, confirms hepatocytes directly contribute to cross-presentation of Ags and priming the pool of naive CD8+ T cells within the liver microenvironment. Processing of soluble and cell-associated Ags into peptide displayed by MHC I is however defective in hepatocytes lacking collectrin, an intracellular chaperone protein that localizes within the endoplasmic reticulum-Golgi intermediate compartment. Loss of hepatic collectrin expression leads to the diminished cross-priming and expansion of cytolytic antiviral CD8+ T cells. This study demonstrates that collectrin positively regulates processing of engulfed Ags into MHC I:peptide complexes within hepatocytes. Collectrin-mediated cross-presentation supports intrahepatic adaptive antiviral immune responses and may lead to insights into the nature of how the liver acts as a primary site of CD8+ T cell activation.


Subject(s)
Adenoviridae Infections/immunology , Adenoviridae/immunology , CD8-Positive T-Lymphocytes/immunology , Cross-Priming , Hepatocytes/immunology , Liver/immunology , Membrane Glycoproteins/metabolism , Acute Disease , Animals , Antigens, Viral/immunology , CD8-Positive T-Lymphocytes/virology , Extracellular Space/immunology , Hepatocytes/virology , Histocompatibility Antigens Class I/metabolism , Liver/virology , Lymphocyte Activation/genetics , Membrane Glycoproteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Solubility , Transplantation Chimera
10.
J Am Soc Nephrol ; 29(1): 155-167, 2018 01.
Article in English | MEDLINE | ID: mdl-28993503

ABSTRACT

Intronic variants of the MYH9 gene that encodes the nonmuscle myosin heavy chain IIA are associated with diabetic nephropathy in European Americans and with sickle cell disease-associated nephropathy. However, the causal functional variants of MYH9 have remained elusive. Rare missense mutations in MYH9 cause macrothrombocytopenia and are occasionally associated with development of nephropathy. The E1841K mutation is among the common MYH9 missense mutations and has been associated with nephropathy in some carriers. To determine the contribution of the E1841K mutation in kidney disease, we studied the effects of the E1841K mutation in mice subjected to high salt or angiotensin II (Ang II) as models of hypertension and in mice subjected to renal mass reduction as a model of CKD. Despite similar levels of BP among wild-type (MYH9+/+ ) mice and mice heterozygous (MYH9+/E1841K ) and homozygous (MYH9E1841K/E1841K ) for the mutation in each model, MYH9E1841K/E1841K mice exhibited mildly increased albuminuria in response to high salt; severe albuminuria, nephrinuria, FSGS, and podocyte foot effacement in Ang II-induced hypertension; and early mortality in the renal mass reduction model. Treatment with candesartan during Ang II-induced hypertension attenuated kidney disease development in MYH9E1841K/E1841K mice. In vitro, isolated primary podocytes from MYH9E1841K/E1841K mice exhibited increased lamellipodia formation and reorganization of F-actin stress fibers. Wound healing assays revealed that MYH9+/+ podocytes had the lowest migration rate, followed by MYH9+/E1841K then MYH9E1841K/E1841K podocytes. In conclusion, the MYH9 E1841K variant alters podocyte cytoskeletal structure and renders podocytes more susceptible to injury after a damaging stimulus.


Subject(s)
Albuminuria/genetics , Cell Movement/genetics , Nonmuscle Myosin Type IIA/genetics , Podocytes/ultrastructure , Renal Insufficiency, Chronic/genetics , Actins/metabolism , Angiotensin II , Animals , Antihypertensive Agents/therapeutic use , Benzimidazoles/therapeutic use , Biphenyl Compounds , Blood Pressure/genetics , Cells, Cultured , Disease Models, Animal , Genotype , Hypertension/chemically induced , Hypertension/genetics , Hypertension/physiopathology , Male , Mice , Mutation, Missense , Myosin Heavy Chains , Nephrectomy , Podocytes/physiology , Primary Cell Culture , Renal Insufficiency, Chronic/pathology , Sodium Chloride, Dietary/administration & dosage , Stress Fibers/genetics , Tetrazoles/therapeutic use
11.
J Am Soc Nephrol ; 28(6): 1826-1837, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28062568

ABSTRACT

Collectrin, encoded by the Tmem27 gene, is a transmembrane glycoprotein with approximately 50% homology with angiotensin converting enzyme 2, but without a catalytic domain. Collectrin is most abundantly expressed in the kidney proximal tubule and collecting duct epithelia, where it has an important role in amino acid transport. Collectrin is also expressed in endothelial cells throughout the vasculature, where it regulates L-arginine uptake. We previously reported that global deletion of collectrin leads to endothelial dysfunction, augmented salt sensitivity, and hypertension. Here, we performed kidney crosstransplants between wild-type (WT) and collectrin knockout (Tmem27Y/- ) mice to delineate the specific contribution of renal versus extrarenal collectrin on BP regulation and salt sensitivity. On a high-salt diet, WT mice with Tmem27Y/- kidneys had the highest systolic BP and were the only group to exhibit glomerular mesangial hypercellularity. Additional studies showed that, on a high-salt diet, Tmem27Y/- mice had lower renal blood flow, higher abundance of renal sodium-hydrogen antiporter 3, and lower lithium clearance than WT mice. In WT mice, administration of angiotensin II for 2 weeks downregulated collectrin expression in a type 1 angiotensin II receptor-dependent manner. This downregulation coincided with the onset of hypertension, such that WT and Tmem27Y/- mice had similar levels of hypertension after 2 weeks of angiotensin II administration. Altogether, these data suggest that salt sensitivity is determined by intrarenal collectrin, and increasing the abundance or activity of collectrin may have therapeutic benefits in the treatment of hypertension and salt sensitivity.


Subject(s)
Angiotensin II/physiology , Down-Regulation , Hypertension/etiology , Membrane Glycoproteins/physiology , Sodium Chloride, Dietary/adverse effects , Animals , Kidney/metabolism , Male , Membrane Glycoproteins/biosynthesis , Mice , Mice, Knockout
13.
Circulation ; 128(16): 1770-80, 2013 Oct 15.
Article in English | MEDLINE | ID: mdl-24048198

ABSTRACT

BACKGROUND: Collectrin is an orphan member of the renin-angiotensin system and is a homolog of angiotensin-converting enzyme 2, sharing ≈50% sequence identity. Unlike angiotensin-converting enzyme 2, collectrin lacks any catalytic domain. Collectrin has been shown to function as a chaperone of amino acid transporters. In rodents, the renal expression of collectrin is increased after subtotal nephrectomy and during high-salt feeding, raising the question of whether collectrin has any direct role in blood pressure regulation. METHODS AND RESULTS: Using a susceptible genetic background, we demonstrate that deletion of collectrin results in hypertension, exaggerated salt sensitivity, and impaired pressure natriuresis. Collectrin knockout mice display impaired endothelium-dependent vasorelaxation that is associated with vascular remodeling, endothelial nitric oxide synthase uncoupling, decreased nitric oxide production, and increased superoxide generation. Treatment with Tempol, a superoxide scavenger, attenuates the augmented sodium sensitivity in collectrin knockout mice. We report for the first time that collectrin is expressed in endothelial cells. Furthermore, collectrin directly regulates l-arginine uptake and plasma membrane levels of CAT1 and y(+)LAT1 amino acid transporters in endothelial cells. Treatment with l-arginine modestly lowers blood pressure of collectrin knockout mice. CONCLUSIONS: Collectrin is a consequential link between the transport of l-arginine and endothelial nitric oxide synthase uncoupling in hypertension.


Subject(s)
Hypertension, Renal/metabolism , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Nitric Oxide Synthase Type III/metabolism , Peptidyl-Dipeptidase A/genetics , Angiotensin-Converting Enzyme 2 , Animals , Arginine/metabolism , Endothelial Cells/cytology , Endothelial Cells/metabolism , Female , Hypertension, Renal/genetics , Hypertension, Renal/physiopathology , Kidney/metabolism , Kidney/physiopathology , Lung/cytology , Male , Mice , Mice, 129 Strain , Mice, Knockout , Natriuresis/physiology , Nitric Oxide/metabolism , Peptidyl-Dipeptidase A/metabolism , Primary Cell Culture , Sodium Chloride, Dietary/pharmacology , Superoxides/metabolism
14.
Am J Physiol Renal Physiol ; 304(4): F348-55, 2013 Feb 15.
Article in English | MEDLINE | ID: mdl-23220723

ABSTRACT

Oxidative stress is acknowledged to play a role in kidney disease progression. Genetic variants that affect the capacity to handle oxidative stress may therefore influence the outcome of kidney disease. We examined whether genetic variants of the GSTM1 gene, a member of a superfamily of glutathione S-transferases, influence the course of kidney disease progression in participants of the African American Study of Kidney Disease (AASK) trial. Groups with and without the common GSTM1 null allele, GSTM1(0), differed significantly in the time to a glomerular filtration rate (GFR) event or dialysis (P = 0.04) and in the time to GFR event, dialysis, or death (P = 0.02). The hazard ratios (HR) for the time to a GFR event or dialysis in those with two or one null allele relative to those possessing none were 1.88 [95% confidence interval (CI), 1.07 to 3.30, P = 0.03] and 1.68 (95% CI, 1.00 to 2.84, P < 0.05), respectively. For the time to GFR event, dialysis, or death, the HR for two null alleles was 2.06 (95% CI, 1.20 to 3.55, P = 0.01) and for one null allele 1.70 (95% CI, 1.02 to 2.81, P = 0.04). We demonstrated that GSTM1 directly regulates intracellular levels of 4-hydroxynonenal (4-HNE) in vascular smooth muscle cells. Furthermore, we showed that renal 4-HNE levels and GSTM1 are both increased after reduction of renal mass (RRM) in the mouse. We conclude that GSTM1 is normally upregulated in chronic kidney disease (CKD) in a protective response to increased oxidative stress. A genetic variant that results in loss of GSTM1 activity may be deleterious in CKD.


Subject(s)
Glutathione Transferase/genetics , Glutathione Transferase/physiology , Hypertension, Renal/ethnology , Hypertension, Renal/genetics , Nephritis/ethnology , Nephritis/genetics , Aldehydes/metabolism , Animals , Black People/genetics , Black People/statistics & numerical data , Cells, Cultured , Disease Progression , Female , Gene Silencing/physiology , Glomerular Filtration Rate/genetics , Glomerular Filtration Rate/physiology , Humans , Hypertension, Renal/enzymology , Hypertension, Renal/mortality , Male , Mice , Mice, Inbred C57BL , Middle Aged , Muscle, Smooth, Vascular/physiology , Myocytes, Smooth Muscle/physiology , NF-E2-Related Factor 2/physiology , Nephritis/enzymology , Nephritis/mortality , Oxidative Stress/genetics , Oxidative Stress/physiology , Randomized Controlled Trials as Topic , Renal Insufficiency, Chronic/enzymology , Renal Insufficiency, Chronic/genetics , Renal Insufficiency, Chronic/physiopathology
15.
Front Pharmacol ; 10: 615, 2019.
Article in English | MEDLINE | ID: mdl-31244655

ABSTRACT

Background: Acute kidney injury (AKI) portends worse prognosis following sepsis, with limited available interventions. Host iron acquisition by pathogens and systemic inflammatory response are key events in the pathogenesis of sepsis. In sepsis, hepcidin induces iron sequestration to limit iron availability to pathogens. Hepcidin is also known to limit inflammation. Since its role in pathophysiology of sepsis-associated AKI is unknown, we investigated the effect of exogenous hepcidin in endotoxin- and peritonitis-induced pathology and AKI. Methods: C57BL/6 mice were treated with saline or 50-100 µg of hepcidin, pre- and post-LPS injection, or cecal ligation and puncture (CLP, model of peritonitis). Splenectomized mice were challenged with LPS, with and without hepcidin. Mice were euthanized at 24 h after LPS injection and at different time points after CLP. Systemic inflammation and renal injury markers were assessed. Direct effect of hepcidin on renal tubular and endothelial cells was evaluated using endotoxin-induced cytotoxic serum. Role of heavy chain ferritin (H-ferritin) in mediating hepcidin-induced anti-inflammatory effect on LPS stimulated macrophages was evaluated with siRNA studies. Results: Twenty-four hours pretreatment with hepcidin significantly reduced LPS-induced AKI. Hepcidin ameliorated LPS-induced increase in serum TNFα and renal Cox-2, and prevented loss in PGC1α and cytochrome c oxidase activity. This was associated with reduced glomerular injury and preserved mitochondrial structure. Hepcidin did not exert direct protection on the renal parenchymal cells but reduced endotoxin-induced serum cytotoxicity to mitigate renal injury. Splenectomy reduced LPS-induced early inflammation and AKI, independent of hepcidin, indicating the importance of systemic inflammation. Higher splenic H-ferritin in hepcidin-treated animals was associated with reduced splenocytes apoptosis and inflammation. Hepcidin reduced LPS-induced IL-6 secretion in macrophages in H-ferritin dependent manner. Hepcidin significantly reduced CLP-induced AKI, and mortality (20% hepcidin treated vs 80% PBS treated). Importantly hepcidin reduced bacteremia and AKI even when administered after onset of sepsis. Conclusion: We demonstrate a protective role of hepcidin in endotoxin- and peritonitis-induced pathologies and AKI, exerted primarily through its anti-inflammatory effects, and antibacterial property. Macrophage H-ferritin plays an important role in hepcidin-mediated protection against endotoxin-induced inflammation. We uncover a novel prophylactic and therapeutic role of hepcidin in sepsis-associated bacteremia, AKI, and mortality.

16.
Hypertension ; 73(2): 407-414, 2019 02.
Article in English | MEDLINE | ID: mdl-30580687

ABSTRACT

The renin-angiotensin system tightly controls aldosterone synthesis. Dysregulation is evident in hypertension (primary aldosteronism), low renin, and resistant hypertension) but also can exist in normotension. Whether chronic, mild aldosterone autonomy can elicit hypertension remains untested. Previously, we reported that global genetic deletion of 2 pore-domain TWIK-relative acid-sensitive potassium channels, TASK-1 and TASK-3, from mice produces striking aldosterone excess, low renin, and hypertension. Here, we deleted TASK-1 and TASK-3 channels selectively from zona glomerulosa cells and generated a model of mild aldosterone autonomy with attendant hypertension that is aldosterone-driven and Ang II (angiotensin II)-independent. This study shows that a zona glomerulosa-specific channel defect can produce mild autonomous hyperaldosteronism sufficient to cause chronic blood pressure elevation.


Subject(s)
Aldosterone/physiology , Angiotensin II/physiology , Hypertension/etiology , Nerve Tissue Proteins/physiology , Potassium Channels, Tandem Pore Domain/physiology , Potassium Channels/physiology , Zona Glomerulosa/physiology , Animals , Male , Mice , Mice, Knockout , Renin-Angiotensin System/physiology
17.
Nat Commun ; 10(1): 29, 2019 01 03.
Article in English | MEDLINE | ID: mdl-30604766

ABSTRACT

Chronic kidney disease (CKD) affects ~10% of the global population, with considerable ethnic differences in prevalence and aetiology. We assemble genome-wide association studies of estimated glomerular filtration rate (eGFR), a measure of kidney function that defines CKD, in 312,468 individuals of diverse ancestry. We identify 127 distinct association signals with homogeneous effects on eGFR across ancestries and enrichment in genomic annotations including kidney-specific histone modifications. Fine-mapping reveals 40 high-confidence variants driving eGFR associations and highlights putative causal genes with cell-type specific expression in glomerulus, and in proximal and distal nephron. Mendelian randomisation supports causal effects of eGFR on overall and cause-specific CKD, kidney stone formation, diastolic blood pressure and hypertension. These results define novel molecular mechanisms and putative causal genes for eGFR, offering insight into clinical outcomes and routes to CKD treatment development.


Subject(s)
Glomerular Filtration Rate/genetics , Hypertension/genetics , Kidney Calculi/genetics , Kidney/physiopathology , Renal Insufficiency, Chronic/genetics , Adult , Aged , Blood Pressure/genetics , Ethnicity/genetics , Female , Genetic Loci/genetics , Genome-Wide Association Study , Histone Code/genetics , Histones/metabolism , Humans , Hypertension/ethnology , Hypertension/physiopathology , Kidney Calculi/ethnology , Kidney Calculi/physiopathology , Male , Middle Aged , Polymorphism, Single Nucleotide , Renal Insufficiency, Chronic/ethnology , Renal Insufficiency, Chronic/physiopathology
18.
J Neurochem ; 105(4): 1253-63, 2008 May.
Article in English | MEDLINE | ID: mdl-18194431

ABSTRACT

Adenosine is an endogenous byproduct of metabolism that regulates cerebral blood flow and modulates neurotransmission. Four receptors, with affinities ranging from nanomolar to micromolar, mediate the effects of adenosine. Real-time measurements are needed to understand the extracellular adenosine concentrations available to activate these receptors. In this study, we measured the subsecond time course of adenosine efflux in the caudate-putamen of anesthetized rats after a 1 s, high-frequency stimulation of dopamine neurons in the substantia nigra. Fast-scan cyclic voltammetry at carbon-fiber microelectrodes was used for simultaneous detection of adenosine and dopamine, which have different oxidation potentials. While dopamine was immediately released after electrical stimulation, adenosine accumulation was slightly delayed and cleared in about 15 seconds. The concentration of adenosine measured after electrical stimulation was 0.94 +/- 0.09 microM. An adenosine kinase inhibitor, adenosine transport inhibitor, and a histamine synthetic precursor were used to pharmacologically confirm the identity of the measured substance as adenosine. Adenosine efflux was also correlated with increases in oxygen, which occur because of changes in cerebral blood flow. This study shows that extracellular adenosine transiently increases after short bursts of neuronal activity in concentrations that can activate receptors.


Subject(s)
Adenosine/metabolism , Caudate Nucleus/metabolism , Putamen/metabolism , Animals , Electric Stimulation/methods , Extracellular Fluid/metabolism , Male , Microelectrodes , Rats , Rats, Sprague-Dawley
19.
J Med Chem ; 46(12): 2494-501, 2003 Jun 05.
Article in English | MEDLINE | ID: mdl-12773053

ABSTRACT

We have recently discovered a novel class of compounds that have oral general anesthetic activity, potent anticonvulsant activity, and minimal hemodynamic effects. The 3,3,3-trifluoro-2-hydroxy-2-phenyl-propionamide (1) demonstrated potent ability to reduce the minimum alveolar concentration (MAC) of isoflurane, with no effects on heart rate or blood pressure at therapeutic concentrations. Analogue 1 also had potent oral anticonvulsant activity against maximal electroshock (MES) and subcutaneous metrazol (scMET) models with a therapeutic index of 10 for MES activity. In this study, we further synthesized nine new racemic analogues and evaluated these compounds for effects on isoflurane MAC reduction and blood pressure. Preliminary data demonstrate potent reduction in the isoflurane MAC for two new compounds. Current mechanistic studies were unrevealing for effects on voltage-gated ion channels as a putative mechanism. Liposomal partitioning studies using (19)F NMR reveal that the aromatic region partitions into the core of the lipid. This partitioning correlated with general anesthetic activity of this class of compounds. Further, compound 1 was used at a concentration of 1 mM and slightly enhanced GABA(A) current in hippocampal neurons at 10 microM. Altogether, 3,3,3-trifluoro-2-hydroxy-2-phenyl-propionamide exhibited excellent oral general anesthetic activity and appears devoid of significant side effects (i.e., alterations in blood pressure or heart rate).


Subject(s)
Amides/chemical synthesis , Anesthetics, General/chemical synthesis , Propionates/chemical synthesis , Administration, Oral , Amides/chemistry , Amides/pharmacology , Anesthetics, General/chemistry , Anesthetics, General/pharmacology , Animals , Anticonvulsants/chemical synthesis , Anticonvulsants/chemistry , Anticonvulsants/pharmacology , Blood Pressure/drug effects , Cells, Cultured , Drug Design , Heart Rate/drug effects , Hippocampus/cytology , Hippocampus/physiology , In Vitro Techniques , Ion Channel Gating , Ion Channels/drug effects , Ion Channels/physiology , Isoflurane/analysis , Liposomes/chemistry , Magnetic Resonance Spectroscopy , Male , Mice , Propionates/chemistry , Propionates/pharmacology , Pulmonary Alveoli/drug effects , Rats , Rats, Sprague-Dawley , Receptors, GABA-A/drug effects , Receptors, GABA-A/physiology , Stereoisomerism , Structure-Activity Relationship , Xenopus
20.
Anesth Analg ; 99(3): 752-757, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15333406

ABSTRACT

Nitric oxide (NO), a cell messenger for activating soluble guanylyl cyclase, is produced by activation of the enzyme NO synthase (NOS) in a wide variety of tissues, including the central nervous system. We have previously demonstrated that inhibition of NOS decreased the minimum alveolar anesthesia concentration (MAC) for isoflurane anesthesia. Moving more distally in the NOS-guanylyl cyclase signaling pathway, we investigated the effects of the specific soluble guanylyl cyclase inhibitor ODQ, 1H-(1,2,4)oxadiazolo[4,3-a]quinoxalin-1-one, on anesthetic requirements. The effect of ODQ on the MAC of isoflurane anesthesia was investigated in Sprague-Dawley rats while concurrently monitoring the their arterial blood pressure and heart rate. After determining control MAC, ODQ 20-500 mg/kg was administered intraperitoneally 30 min before re-determining MAC in the presence of the soluble guanylyl cyclase inhibitor. In one series, the effect of 250 mg/kg of ODQ on neuronal cyclase guanosine monophosphate production was determined by microdialysis. ODQ produced a statistically significant, dose-dependent decrease from isoflurane control MAC (maximal effect 52.4% +/- 2.7%). No ceiling effect was observed over the dose-range studied. This reduction in isoflurane MAC was not accompanied by changes in either heart rate or blood pressure. Inhibition of the NOS-guanylyl cyclase signaling pathway decreased the MAC for isoflurane, which suggests that inhibition of this pathway may play a role in the anesthetic state. The MAC reduction by the soluble guanylyl cyclase inhibitor ODQ was devoid of any significant hemodynamic effects. The current findings, along with the previous observations that structurally distinct NOS inhibitors and the nonspecific soluble guanylyl cyclase inhibitor methylene blue decrease the MAC for volatile anesthetics, support that this is an effect specific to the NOS-guanylyl cyclase signaling pathway.


Subject(s)
Anesthetics, Inhalation/pharmacology , Enzyme Inhibitors/pharmacology , Guanylate Cyclase/antagonists & inhibitors , Isoflurane/pharmacology , Oxadiazoles/pharmacology , Quinoxalines/pharmacology , Animals , Dose-Response Relationship, Drug , Guanylate Cyclase/physiology , Male , N-Methylaspartate/pharmacology , Nitric Oxide Synthase/physiology , Rats , Rats, Sprague-Dawley , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL