Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 142
Filter
1.
Hypertension ; 81(4): 682-686, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38507510

ABSTRACT

Renin was discovered more than a century ago. Since then, the functions of the renin-angiotensin system in the kidney have been the focus of intensive research revealing its importance in regulation of renal physiology and in the pathogenesis of heart, vascular, and kidney diseases. Inhibitors of renin-angiotensin system components are now foundational therapies for a range of kidney and cardiovascular diseases from hypertension to heart failure to diabetic nephropathy. Despite years of voluminous research, emerging studies continue to reveal new complexities of the regulation of the renin-angiotensin system within the kidney and identification of nonclassical components of the system like the prorenin receptor (PRR) and ACE2 (angiotensin-converting enzyme 2), with powerful renal effects that ultimately impact the broader cardiovascular system. With the emergence of a range of novel therapies for cardiovascular and kidney diseases, the importance of a detailed understanding of the renin-angiotensin system in the kidney will allow for the development of informed complementary approaches for combinations of treatments that will optimally promote health and longevity over the century ahead.


Subject(s)
Diabetic Nephropathies , Hypertension , Humans , Renin-Angiotensin System , Health Promotion , Kidney/metabolism , Renin/metabolism , Diabetic Nephropathies/metabolism
2.
Article in English | MEDLINE | ID: mdl-38546133

ABSTRACT

CONTEXT: Metabolites in tricarboxylic acid (TCA) pathway have pleiotropic functions. OBJECTIVE: To study the association between urine TCA cycle metabolites and the risk for chronic kidney disease (CKD) progression in individuals with type 2 diabetes. DESIGN, SETTING AND PARTICIPANTS: A prospective study in a discovery (n = 1826) and a validation (n = 1235) cohort of type 2 diabetes in a regional hospital and a primary care facility. EXPOSURE AND OUTCOME: Urine lactate, pyruvate, citrate, alpha-ketoglutarate, succinate, fumarate and malate were measured by mass spectrometry. CKD progression was defined as a composite of sustained eGFR below 15 ml/min/1.73 m2 , dialysis, renal death or doubling of serum creatinine. RESULTS: During a median of 9.2 (IQR 8.1-9.7) and 4.0 (3.2-5.1) years of follow-up, 213 and 107 renal events were identified. Cox regression suggested that urine lactate, fumarate and malate were associated with an increased risk (adjusted hazard ratio, aHR [95% CI] 1.63 [1.16-2.28], 1.82 [1.17-2.82] and 1.49 [1.05-2.11], per SD), while citrate was associated with a low risk (aHR 0.83 [0.72-0.96] per SD) for the renal outcome after adjustment for cardio-renal risk factors. These findings were reproducible in the validation cohort. Noteworthy, fumarate and citrate were independently associated with the renal outcome after additional adjustment for other metabolites. CONCLUSION: Urine fumarate and citrate predict the risk for progression to ESKD independent of clinical risk factors and other urine metabolites. These two metabolites in TCA cycle pathway may play important roles in the pathophysiological network underpinning progressive loss of kidney function in patients with type 2 diabetes.

3.
J Clin Invest ; 134(9)2024 Mar 14.
Article in English | MEDLINE | ID: mdl-38483511

ABSTRACT

In lung, thromboxane A2 (TXA2) activates the TP receptor to induce proinflammatory and bronchoconstrictor effects. Thus, TP receptor antagonists and TXA2 synthase inhibitors have been tested as potential asthma therapeutics in humans. Th9 cells play key roles in asthma and regulate the lung immune response to allergens. Herein, we found that TXA2 reduces Th9 cell differentiation during allergic lung inflammation. Th9 cells were decreased approximately 2-fold and airway hyperresponsiveness was attenuated in lungs of allergic mice treated with TXA2. Naive CD4+ T cell differentiation to Th9 cells and IL-9 production were inhibited dose-dependently by TXA2 in vitro. TP receptor-deficient mice had an approximately 2-fold increase in numbers of Th9 cells in lungs in vivo after OVA exposure compared with wild-type mice. Naive CD4+ T cells from TP-deficient mice exhibited increased Th9 cell differentiation and IL-9 production in vitro compared with CD4+ T cells from wild-type mice. TXA2 also suppressed Th2 and enhanced Treg differentiation both in vitro and in vivo. Thus, in contrast to its acute, proinflammatory effects, TXA2 also has longer-lasting immunosuppressive effects that attenuate the Th9 differentiation that drives asthma progression. These findings may explain the paradoxical failure of anti-thromboxane therapies in the treatment of asthma.


Subject(s)
Asthma , Cell Differentiation , T-Lymphocytes, Regulatory , Th2 Cells , Thromboxane A2 , Animals , Mice , Th2 Cells/immunology , Th2 Cells/pathology , Thromboxane A2/metabolism , Thromboxane A2/immunology , T-Lymphocytes, Regulatory/immunology , Asthma/immunology , Asthma/pathology , Asthma/drug therapy , Asthma/genetics , Mice, Knockout , Interleukin-9/immunology , Interleukin-9/genetics , Interleukin-9/metabolism , Pneumonia/immunology , Pneumonia/pathology , Mice, Inbred C57BL , Mice, Inbred BALB C , Lung/immunology , Lung/pathology , Ovalbumin/immunology , Female , T-Lymphocytes, Helper-Inducer/immunology
4.
BMC Nephrol ; 25(1): 23, 2024 Jan 17.
Article in English | MEDLINE | ID: mdl-38233790

ABSTRACT

BACKGROUND: We aimed to explore the three-way interaction among age, gender, and kidney function on the risk of all-cause mortality and cardiovascular mortality among patients with type 2 diabetes (T2D). METHODS: In a retrospective cohort study, patients aged > 40 years with T2D with serum creatinine and urine albumin measured from 2013 to 2019 were included from a multi-institutional diabetes registry. The exposure was estimated glomerular filtration rate (eGFR), outcomes were all-cause mortality (primary outcome) and cardiovascular disease (CVD) mortality (secondary outcome). We applied multivariable cox proportional hazards regression analysis to compute the association between eGFR and mortality. RESULTS: A total of 36,556 patients were followed for up to 6 years during which 2492 (6.82%) died from all causes, and 690 (1.9%) died from CVD. We observed a significant three-way interaction (p = 0.021) among age (younger, < 65; older, ≥65 years), gender and eGFR for the risk of all-cause mortality. Using age- and gender-specific eGFR of 90 ml/min/1.73m2 as the reference point, the adjusted hazard rate (HR) (95% CI) for all-cause mortality at eGFR of 40 ml/min/1.73m2 was 3.70 (2.29 to 5.99) in younger women and 1.86 (1.08 to 3.19) in younger men. The corresponding adjusted HRs in older women and older men were 2.38 (2.02 to 2.82) and 2.18 (1.85 to 2.57), respectively. Similar results were observed for CVD deaths, although the three-way interaction was not statistically significant. Sensitivity analysis yielded similar results. CONCLUSIONS: In this T2D population, younger women with reduced kidney function might be more susceptible to higher risks of all-cause mortality and CVD mortality than younger men.


Subject(s)
Cardiovascular Diseases , Diabetes Mellitus, Type 2 , Male , Humans , Female , Aged , Cohort Studies , Retrospective Studies , Singapore , Glomerular Filtration Rate , Kidney , Registries , Risk Factors
5.
Kidney Int ; 104(6): 1135-1149, 2023 12.
Article in English | MEDLINE | ID: mdl-37843477

ABSTRACT

Diabetic nephropathy (DN) is characterized by abnormal kidney energy metabolism, but its causes and contributions to DN pathogenesis are not clear. To examine this issue, we carried out targeted metabolomics profiling in a mouse model of DN that develops kidney disease resembling the human disorder. We found a distinct profile of increased lactate levels and impaired energy metabolism in kidneys of mice with DN, and treatment with an angiotensin-receptor blocker (ARB) reduced albuminuria, attenuated kidney pathology and corrected many metabolic abnormalities, restoring levels of lactate toward normal while increasing kidney ATP content. We also found enhanced expression of lactate dehydrogenase isoforms in DN. Expression of both the LdhA and LdhB isoforms were significantly increased in kidneys of mice, and treatment with ARB significantly reduced their expression. Single-cell sequencing studies showed specific up-regulation of LdhA in the proximal tubule, along with enhanced expression of oxidative stress pathways. There was a significant correlation between albuminuria and lactate in mice, and also in a Southeast Asian patient cohort consisting of individuals with type 2 diabetes and impaired kidney function. In the individuals with diabetes, this association was independent of ARB and angiotensin-converting enzyme inhibitor use. Furthermore, urinary lactate levels predicted the clinical outcomes of doubling of serum creatinine or development of kidney failure, and there was a significant correlation between urinary lactate levels and biomarkers of tubular injury and epithelial stress. Thus, we suggest that kidney metabolic disruptions leading to enhanced generation of lactate contribute to the pathogenesis of DN and increased urinary lactate levels may be a potential biomarker for risk of kidney disease progression.


Subject(s)
Diabetes Mellitus, Type 2 , Diabetic Nephropathies , Renal Insufficiency , Humans , Animals , Mice , Diabetic Nephropathies/etiology , Lactic Acid , Albuminuria/etiology , Angiotensin Receptor Antagonists/pharmacology , Angiotensin Receptor Antagonists/therapeutic use , Diabetes Mellitus, Type 2/complications , Angiotensin-Converting Enzyme Inhibitors/pharmacology , Angiotensin-Converting Enzyme Inhibitors/therapeutic use , Kidney , Protein Isoforms
6.
Hypertension ; 80(3): 668-677, 2023 03.
Article in English | MEDLINE | ID: mdl-36628961

ABSTRACT

BACKGROUND: Ang II (angiotensin II) type 1 (AT1) receptors play a critical role in cardiovascular diseases such as hypertension. Rodents have 2 types of AT1 receptor (AT1A and AT1B) of which knock-in Tagln-mediated smooth muscle AT1A silencing attenuated Ang II-induced hypertension. Although vascular remodeling, a significant contributor to organ damage, occurs concurrently with hypertension in Ang II-infused mice, the contribution of smooth muscle AT1A in this process remains unexplored. Accordingly, it is hypothesized that smooth muscle AT1A receptors exclusively contribute to both medial thickening and adventitial fibrosis regardless of the presence of hypertension. METHODS: About 1 µg/kg per minute Ang II was infused for 2 weeks in 2 distinct AT1A receptor silenced mice, knock-in Tagln-mediated constitutive smooth muscle AT1A receptor silenced mice, and Myh11-mediated inducible smooth muscle AT1A together with global AT1B silenced mice for evaluation of hypertensive cardiovascular remodeling. RESULTS: Medial thickness, adventitial collagen deposition, and immune cell infiltration in aorta were increased in control mice but not in both smooth muscle AT1A receptor silenced mice. Coronary arterial perivascular fibrosis in response to Ang II infusion was also attenuated in both AT1A receptor silenced mice. Ang II-induced cardiac hypertrophy was attenuated in constitutive smooth muscle AT1A receptor silenced mice. However, Ang II-induced cardiac hypertrophy and hypertension were not altered in inducible smooth muscle AT1A receptor silenced mice. CONCLUSIONS: Smooth muscle AT1A receptors mediate Ang II-induced vascular remodeling including medial hypertrophy and inflammatory perivascular fibrosis regardless of the presence of hypertension. Our data suggest an independent etiology of blood pressure elevation and hypertensive vascular remodeling in response to Ang II.


Subject(s)
Hypertension , Receptor, Angiotensin, Type 1 , Mice , Animals , Receptor, Angiotensin, Type 1/genetics , Angiotensin II/pharmacology , Vascular Remodeling , Myocytes, Smooth Muscle , Cardiomegaly , Fibrosis , Mice, Knockout , Mice, Inbred C57BL
7.
Diabetes ; 72(7): 932-946, 2023 07 01.
Article in English | MEDLINE | ID: mdl-36445944

ABSTRACT

Diabetic corneal neuropathy (DCN) is a common complication of diabetes. However, there are very limited therapeutic options. We investigated the effects of a peroxisome proliferator-activated receptor-α (PPAR-α) agonist, fenofibrate, on 30 patients (60 eyes) with type 2 diabetes. On in vivo confocal microscopy evaluation, there was significant stimulation of corneal nerve regeneration and a reduction in nerve edema after 30 days of oral fenofibrate treatment, as evidenced by significant improvement in corneal nerve fiber density (CNFD) and corneal nerve fiber width, respectively. Corneal epithelial cell morphology also significantly improved in cell circularity. Upon clinical examination, fenofibrate significantly improved patients' neuropathic ocular surface status by increasing tear breakup time along with a reduction of corneal and conjunctival punctate keratopathy. Tear substance P (SP) concentrations significantly increased after treatment, suggesting an amelioration of ocular surface neuroinflammation. The changes in tear SP concentrations was also significantly associated with improvement in CNFD. Quantitative proteomic analysis demonstrated that fenofibrate significantly upregulated and modulated the neurotrophin signaling pathway and linolenic acid, cholesterol, and fat metabolism. Complement cascades, neutrophil reactions, and platelet activation were also significantly suppressed. Our results showed that fenofibrate could potentially be a novel treatment for patients with DCN.


Subject(s)
Diabetes Mellitus, Type 2 , Diabetic Neuropathies , Fenofibrate , Humans , PPAR alpha/metabolism , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/drug therapy , Fenofibrate/pharmacology , Fenofibrate/therapeutic use , Proteomics , Cornea/innervation , Hypoglycemic Agents , Diabetic Neuropathies/drug therapy , Diabetic Neuropathies/diagnosis , Microscopy, Confocal/methods
8.
Nat Commun ; 13(1): 7497, 2022 12 05.
Article in English | MEDLINE | ID: mdl-36470928

ABSTRACT

The kidney has large regenerative capacity, but this is compromised when kidney damage is excessive and renal tubular epithelial cells (TECs) undergo SNAI1-driven growth arrest. Here we investigate the role of IL11 in TECs, kidney injury and renal repair. IL11 stimulation of TECs induces ERK- and p90RSK-mediated GSK3ß inactivation, SNAI1 upregulation and pro-inflammatory gene expression. Mice with acute kidney injury upregulate IL11 in TECs leading to SNAI1 expression and kidney dysfunction, which is not seen in Il11 deleted mice or in mice administered a neutralizing IL11 antibody in either preemptive or treatment modes. In acute kidney injury, anti-TGFß reduces renal fibrosis but exacerbates inflammation and tubule damage whereas anti-IL11 reduces all pathologies. Mice with TEC-specific deletion of Il11ra1 have reduced pathogenic signaling and are protected from renal injury-induced inflammation, fibrosis, and failure. In a model of chronic kidney disease, anti-IL11 therapy promotes TEC proliferation and parenchymal regeneration, reverses fibroinflammation and restores renal mass and function. These data highlight IL11-induced mesenchymal transition of injured TECs as an important renal pathology and suggest IL11 as a therapeutic target for restoring stalled endogenous regeneration in the diseased kidney.


Subject(s)
Acute Kidney Injury , Antibodies, Neutralizing , Interleukin-11 , Kidney Tubules , Nephritis , Regeneration , Renal Insufficiency, Chronic , Animals , Mice , Acute Kidney Injury/therapy , Fibrosis , Interleukin-11 Receptor alpha Subunit/genetics , Kidney Tubules/physiology , Nephritis/therapy , Interleukin-11/antagonists & inhibitors , Interleukin-11/physiology , Gene Deletion , Antibodies, Neutralizing/therapeutic use , Renal Insufficiency, Chronic/therapy , Disease Models, Animal
9.
J Am Heart Assoc ; 11(19): e026581, 2022 10 04.
Article in English | MEDLINE | ID: mdl-36172956

ABSTRACT

Background A beneficial role for prostanoids in hypertension is suggested by clinical studies showing nonsteroidal anti-inflammatory drugs, which block the production of all prostanoids, cause sodium retention and exacerbate hypertension. Among prostanoids, prostaglandin E2 and its E-prostanoid receptor 4 receptor (EP4R) have been implicated in blood pressure control. Our previous study found that conditional deletion of EP4R from all tissues in adult mice exacerbates angiotensin II-dependent hypertension, suggesting a powerful effect of EP4R to resist blood pressure elevation. We also found that elimination of EP4R from vascular smooth muscle cells did not affect the severity of hypertension, suggesting nonvascular targets of prostaglandin E mediate this antihypertensive effect. Methods and Results Here we generated mice with cell-specific deletion of EP4R from macrophage-specific EP4 receptor knockouts or kidney epithelial cells (KEKO) to assess the contributions of EP4R in these cells to hypertension pathogenesis. Macrophage-specific EP4 receptor knockouts showed similar blood pressure responses to alterations in dietary sodium or chronic angiotensin II infusion as Controls. By contrast, angiotensin II-dependent hypertension was significantly augmented in KEKOs (mean arterial pressure: 146±3 mm Hg) compared with Controls (137±4 mm Hg; P=0.02), which was accompanied by impaired natriuresis in KEKOs. Because EP4R expression in the kidney is enriched in the collecting duct, we compared responses to amiloride in angiotensin II-infused KEKOs and Controls. Blockade of the epithelial sodium channel with amiloride caused exaggerated natriuresis in KEKOs compared with Controls (0.21±0.01 versus 0.15±0.02 mmol/24 hour per 20 g; P=0.015). Conclusions Our data suggest EP4R in kidney epithelia attenuates hypertension. This antihypertension effect of EP4R may be mediated by reducing the activity of the epithelial sodium channel, thereby promoting natriuresis.


Subject(s)
Hypertension , Receptors, Prostaglandin E, EP4 Subtype , Amiloride/therapeutic use , Angiotensin II/metabolism , Animals , Anti-Inflammatory Agents/therapeutic use , Antihypertensive Agents/therapeutic use , Dinoprostone/metabolism , Epithelial Cells , Epithelial Sodium Channels/genetics , Hypertension/drug therapy , Kidney , Macrophages/metabolism , Mice , Prostaglandins , Receptors, Prostaglandin E, EP4 Subtype/genetics , Receptors, Prostaglandin E, EP4 Subtype/metabolism , Sodium/metabolism , Sodium Chloride, Dietary/metabolism
10.
Kidney360 ; 3(12): 2086-2094, 2022 12 29.
Article in English | MEDLINE | ID: mdl-36591353

ABSTRACT

Background: ACE2 is a key enzyme in the renin-angiotensin system (RAS) capable of balancing the RAS by metabolizing angiotensin II (AngII). First described in cardiac tissue, abundance of ACE2 is highest in the kidney, and it is also expressed in several extrarenal tissues. Previously, we reported an association between enhanced susceptibility to hypertension and elevated renal AngII levels in global ACE2-knockout mice. Methods: To examine the effect of ACE2 expressed in the kidney, relative to extrarenal expression, on the development of hypertension, we used a kidney crosstransplantation strategy with ACE2-KO and WT mice. In this model, both native kidneys are removed and renal function is provided entirely by the transplanted kidney, such that four experimental groups with restricted ACE2 expression are generated: WT→WT (WT), KO→WT (KidneyKO), WT→KO (SystemicKO), and KO→KO (TotalKO). Additionally, we used nanoscale mass spectrometry-based proteomics to identify ACE2 fragments in early glomerular filtrate of mice. Results: Although significant differences in BP were not detected, a major finding of our study is that shed or soluble ACE2 (sACE2) was present in urine of KidneyKO mice that lack renal ACE2 expression. Detection of sACE2 in the urine of KidneyKO mice during AngII-mediated hypertension suggests that sACE2 originating from extrarenal tissues can reach the kidney and be excreted in urine. To confirm glomerular filtration of ACE2, we used micropuncture and nanoscale proteomics to detect peptides derived from ACE2 in the Bowman's space. Conclusions: Our findings suggest that both systemic and renal tissues may contribute to sACE2 in urine, identifying the kidney as a major site for ACE2 actions. Moreover, filtration of sACE2 into the lumen of the nephron may contribute to the pathophysiology of kidney diseases characterized by disruption of the glomerular filtration barrier.


Subject(s)
Angiotensin-Converting Enzyme 2 , Hypertension , Kidney , Renin-Angiotensin System , Animals , Mice , Angiotensin II/metabolism , Angiotensin II/pharmacology , Angiotensin-Converting Enzyme 2/genetics , Angiotensin-Converting Enzyme 2/metabolism , Hypertension/genetics , Hypertension/metabolism , Kidney/metabolism , Mice, Knockout , Peptidyl-Dipeptidase A/genetics , Peptidyl-Dipeptidase A/metabolism , Peptidyl-Dipeptidase A/pharmacology , Renin-Angiotensin System/genetics , Renin-Angiotensin System/physiology
11.
Nat Genet ; 53(9): 1322-1333, 2021 09.
Article in English | MEDLINE | ID: mdl-34385711

ABSTRACT

The functional interpretation of genome-wide association studies (GWAS) is challenging due to the cell-type-dependent influences of genetic variants. Here, we generated comprehensive maps of expression quantitative trait loci (eQTLs) for 659 microdissected human kidney samples and identified cell-type-eQTLs by mapping interactions between cell type abundances and genotypes. By partitioning heritability using stratified linkage disequilibrium score regression to integrate GWAS with single-cell RNA sequencing and single-nucleus assay for transposase-accessible chromatin with high-throughput sequencing data, we prioritized proximal tubules for kidney function and endothelial cells and distal tubule segments for blood pressure pathogenesis. Bayesian colocalization analysis nominated more than 200 genes for kidney function and hypertension. Our study clarifies the mechanism of commonly used antihypertensive and renal-protective drugs and identifies drug repurposing opportunities for kidney disease.


Subject(s)
Hypertension/genetics , Kidney Tubules, Distal/pathology , Kidney Tubules, Proximal/pathology , Quantitative Trait Loci/genetics , Renal Insufficiency, Chronic/genetics , Base Sequence , Chromosome Mapping , Endothelial Cells/pathology , Genetic Predisposition to Disease/genetics , Genome-Wide Association Study , Genotype , High-Throughput Nucleotide Sequencing , Humans , Polymorphism, Single Nucleotide/genetics , Quantitative Trait, Heritable , Renal Insufficiency, Chronic/pathology , Sequence Analysis, RNA , Single-Cell Analysis
12.
Am J Physiol Renal Physiol ; 320(6): F1080-F1092, 2021 06 01.
Article in English | MEDLINE | ID: mdl-33969697

ABSTRACT

A major pathway in hypertension pathogenesis involves direct activation of ANG II type 1 (AT1) receptors in the kidney, stimulating Na+ reabsorption. AT1 receptors in tubular epithelia control expression and stimulation of Na+ transporters and channels. Recently, we found reduced blood pressure and enhanced natriuresis in mice with cell-specific deletion of AT1 receptors in smooth muscle (SMKO mice). Although impaired vasoconstriction and preserved renal blood flow might contribute to exaggerated urinary Na+ excretion in SMKO mice, we considered whether alterations in Na+ transporter expression might also play a role; therefore, we carried out proteomic analysis of key Na+ transporters and associated proteins. Here, we show that levels of Na+-K+-2Cl- cotransporter isoform 2 (NKCC2) and Na+/H+ exchanger isoform 3 (NHE3) are reduced at baseline in SMKO mice, accompanied by attenuated natriuretic and diuretic responses to furosemide. During ANG II hypertension, we found widespread remodeling of transporter expression in wild-type mice with significant increases in the levels of total NaCl cotransporter, phosphorylated NaCl cotransporter (Ser71), and phosphorylated NKCC2, along with the cleaved, activated forms of the α- and γ-epithelial Na+ channel. However, the increases in α- and γ-epithelial Na+ channel with ANG II were substantially attenuated in SMKO mice. This was accompanied by a reduced natriuretic response to amiloride. Thus, enhanced urinary Na+ excretion observed after cell-specific deletion of AT1 receptors from smooth muscle cells is associated with altered Na+ transporter abundance across epithelia in multiple nephron segments. These findings suggest a system of vascular-epithelial in the kidney, modulating the expression of Na+ transporters and contributing to the regulation of pressure natriuresis.NEW & NOTEWORTHY The use of drugs to block the renin-angiotensin system to reduce blood pressure is common. However, the precise mechanism for how these medications control blood pressure is incompletely understood. Here, we show that mice lacking angiotensin receptors specifically in smooth muscle cells lead to alternation in tubular transporter amount and function. Thus, demonstrating the importance of vascular-tubular cross talk in the control of blood pressure.


Subject(s)
Angiotensin II/pharmacology , Epithelial Cells/metabolism , Kidney/blood supply , Myocytes, Smooth Muscle/metabolism , Receptor, Angiotensin, Type 1/metabolism , Amiloride/pharmacology , Animals , Epithelial Sodium Channel Blockers/pharmacology , Female , Furosemide/pharmacology , Gene Expression Regulation/drug effects , Green Fluorescent Proteins , Hypertension/chemically induced , Luminescent Proteins , Male , Mice , Mice, Inbred Strains , Mice, Knockout , Receptor, Angiotensin, Type 1/genetics , Sodium/metabolism , Sodium Potassium Chloride Symporter Inhibitors/pharmacology , Red Fluorescent Protein
13.
Circ Res ; 128(7): 847-863, 2021 04 02.
Article in English | MEDLINE | ID: mdl-33793328

ABSTRACT

Dr Irvine Page proposed the Mosaic Theory of Hypertension in the 1940s advocating that hypertension is the result of many factors that interact to raise blood pressure and cause end-organ damage. Over the years, Dr Page modified his paradigm, and new concepts regarding oxidative stress, inflammation, genetics, sodium homeostasis, and the microbiome have arisen that allow further refinements of the Mosaic Theory. A constant feature of this approach to understanding hypertension is that the various nodes are interdependent and that these almost certainly vary between experimental models and between individuals with hypertension. This review discusses these new concepts and provides an introduction to other reviews in this compendium of Circulation Research.


Subject(s)
Hypertension/physiopathology , Aldosterone/physiology , Angiotensin II/pharmacology , Blood Pressure/drug effects , Blood Pressure/physiology , Blood Vessels/physiology , Body Fluids/physiology , Central Nervous System/physiology , Humans , Hypertension/etiology , Inflammation/complications , Kidney/physiology , Microbiota/physiology , Reactive Oxygen Species/metabolism , Risk Factors , Sodium Chloride/administration & dosage , Sodium Chloride/adverse effects , Vasoconstrictor Agents/pharmacology
14.
Hypertension ; 77(2): 393-404, 2021 02.
Article in English | MEDLINE | ID: mdl-33390039

ABSTRACT

Activation of AT1 (type 1 Ang) receptors stimulates cardiomyocyte hypertrophy in vitro. Accordingly, it has been suggested that regression of cardiac hypertrophy associated with renin-Ang system blockade is due to inhibition of cellular actions of Ang II in the heart, above and beyond their effects to reduce pressure overload. We generated 2 distinct mouse lines with cell-specific deletion of AT1A receptors, from cardiomyocytes. In the first line (C-SMKO), elimination of AT1A receptors was achieved using a heterologous Cre recombinase transgene under control of the Sm22 promoter, which expresses in cells of smooth muscle lineage including cardiomyocytes and vascular smooth muscle cells of conduit but not resistance vessels. The second line (R-SMKO) utilized a Cre transgene knocked-in to the Sm22 locus, which drives expression in cardiac myocytes and vascular smooth muscle cells in both conduit and resistance arteries. Thus, although both groups lack AT1 receptors in the cardiomyocytes, they are distinguished by presence (C-SMKO) or absence (R-SMKO) of peripheral vascular responses to Ang II. Similar to wild-types, chronic Ang II infusion caused hypertension and cardiac hypertrophy in C-SMKO mice, whereas both hypertension and cardiac hypertrophy were reduced in R-SMKOs. Thus, despite the absence of AT1A receptors in cardiomyocytes, C-SMKOs develop robust cardiac hypertrophy. By contrast, R-SMKOs developed identical levels of hypertrophy in response to pressure overload-induced by transverse aortic banding. Our findings suggest that direct activation of AT1 receptors in cardiac myocytes has minimal influence on cardiac hypertrophy induced by renin-Ang system activation or pressure overload.


Subject(s)
Angiotensin II/pharmacology , Cardiomegaly/genetics , Hypertension/genetics , Myocytes, Cardiac/metabolism , Receptor, Angiotensin, Type 1/genetics , Renin-Angiotensin System/drug effects , Animals , Cardiomegaly/chemically induced , Cardiomegaly/metabolism , Hypertension/chemically induced , Hypertension/metabolism , Mice , Mice, Knockout , Mice, Transgenic , Myocardium/metabolism , Myocytes, Cardiac/drug effects , Vascular Resistance/drug effects
16.
Hypertension ; 73(6): e87-e120, 2019 06.
Article in English | MEDLINE | ID: mdl-30866654

ABSTRACT

Hypertension is the most common chronic disease in the world, yet the precise cause of elevated blood pressure often cannot be determined. Animal models have been useful for unraveling the pathogenesis of hypertension and for testing novel therapeutic strategies. The utility of animal models for improving the understanding of the pathogenesis, prevention, and treatment of hypertension and its comorbidities depends on their validity for representing human forms of hypertension, including responses to therapy, and on the quality of studies in those models (such as reproducibility and experimental design). Important unmet needs in this field include the development of models that mimic the discrete hypertensive syndromes that now populate the clinic, resolution of ongoing controversies in the pathogenesis of hypertension, and the development of new avenues for preventing and treating hypertension and its complications. Animal models may indeed be useful for addressing these unmet needs.


Subject(s)
American Heart Association , Antihypertensive Agents/therapeutic use , Biomedical Research , Blood Pressure/physiology , Hypertension/physiopathology , Animals , Disease Models, Animal , Hypertension/drug therapy , United States
17.
J Am Heart Assoc ; 8(4): e010745, 2019 02 19.
Article in English | MEDLINE | ID: mdl-30764697

ABSTRACT

Background Prostaglandin E2 ( PGE 2) is a major prostanoid with multiple actions that potentially affect blood pressure ( BP ). PGE 2 acts through 4 distinct E-prostanoid ( EP ) receptor isoforms: EP 1 to EP 4. The EP 4 receptor ( EP 4R) promotes PGE 2-dependent vasodilation, but its role in the pathogenesis of hypertension is not clear. Methods and Results To address this issue, we studied mice after temporal- and cell-specific deletion of EP 4R. First, using a mouse line with loss of EP 4 expression induced universally after birth, we confirm that EP 4R mediates a major portion of the acute vasodilatory effects of infused PGE 2. In addition, EP 4 contributes to control of resting BP , which was increased by 5±1 mm Hg in animals with generalized deficiency of this receptor. We also show that EP 4 is critical for limiting elevations in BP caused by high salt feeding and long-term infusion of angiotensin II . To more precisely identify the mechanism for these actions, we generated mice in which EP 4R loss is induced after birth and is limited to smooth muscle. In these mice, acute PGE 2-dependent vasodilation was attenuated, indicating that this response is mediated by EP 4R in vascular smooth muscle cells. However, absence of EP 4R only in this vascular compartment had a paradoxical effect of lowering resting BP , whereas the protective effect of EP 4R on limiting angiotensin II-dependent hypertension was unaffected. Conclusions Taken together, our findings support a complex role for EP 4R in regulation of BP and in hypertension, which appears to involve actions of the EP 4R in tissues beyond vascular smooth muscle cells.


Subject(s)
Blood Pressure/physiology , Hypertension/metabolism , Receptors, Prostaglandin E, EP4 Subtype/metabolism , Vasodilation/physiology , Angiotensin II/administration & dosage , Animals , Dinoprostone/administration & dosage , Disease Models, Animal , Hypertension/drug therapy , Hypertension/physiopathology , Infusions, Intravenous , Mice , Mice, Transgenic , Rest , Signal Transduction , Vasoconstrictor Agents/administration & dosage , Vasodilation/drug effects
18.
Diabetes ; 67(10): 2096-2106, 2018 10.
Article in English | MEDLINE | ID: mdl-30065034

ABSTRACT

Diabetic nephropathy (DN) is a leading cause of end-stage renal disease worldwide, but its molecular pathogenesis is not well defined, and there are no specific treatments. In humans, there is a strong genetic component determining susceptibility to DN. However, specific genes controlling DN susceptibility in humans have not been identified. In this study, we describe a mouse model combining type 1 diabetes with activation of the renin-angiotensin system (RAS), which develops robust kidney disease with features resembling human DN: heavy albuminuria, hypertension, and glomerulosclerosis. Additionally, there is a powerful effect of genetic background regulating susceptibility to nephropathy; the 129 strain is susceptible to kidney disease, whereas the C57BL/6 strain is resistant. To examine the molecular basis of this differential susceptibility, we analyzed the glomerular transcriptome of young mice early in the course of their disease. We find dramatic differences in regulation of immune and inflammatory pathways, with upregulation of proinflammatory pathways in the susceptible (129) strain and coordinate downregulation in the resistant (C57BL/6) strain. Many of these pathways are also upregulated in rat models and in humans with DN. Our studies suggest that genes controlling inflammatory responses, triggered by hyperglycemia and RAS activation, may be critical early determinants of susceptibility to DN.


Subject(s)
Diabetic Nephropathies/genetics , Diabetic Nephropathies/immunology , Inflammation/genetics , Inflammation/immunology , Kidney Diseases/genetics , Kidney Diseases/immunology , Animals , Blood Glucose/genetics , Blood Glucose/immunology , Blotting, Western , Genetic Predisposition to Disease/genetics , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction
19.
Physiol Rev ; 98(3): 1627-1738, 2018 07 01.
Article in English | MEDLINE | ID: mdl-29873596

ABSTRACT

The renin-angiotensin-aldosterone system plays crucial roles in cardiovascular physiology and pathophysiology. However, many of the signaling mechanisms have been unclear. The angiotensin II (ANG II) type 1 receptor (AT1R) is believed to mediate most functions of ANG II in the system. AT1R utilizes various signal transduction cascades causing hypertension, cardiovascular remodeling, and end organ damage. Moreover, functional cross-talk between AT1R signaling pathways and other signaling pathways have been recognized. Accumulating evidence reveals the complexity of ANG II signal transduction in pathophysiology of the vasculature, heart, kidney, and brain, as well as several pathophysiological features, including inflammation, metabolic dysfunction, and aging. In this review, we provide a comprehensive update of the ANG II receptor signaling events and their functional significances for potential translation into therapeutic strategies. AT1R remains central to the system in mediating physiological and pathophysiological functions of ANG II, and participation of specific signaling pathways becomes much clearer. There are still certain limitations and many controversies, and several noteworthy new concepts require further support. However, it is expected that rigorous translational research of the ANG II signaling pathways including those in large animals and humans will contribute to establishing effective new therapies against various diseases.


Subject(s)
Angiotensin II/metabolism , Receptors, Angiotensin/metabolism , Signal Transduction , Adipocytes/metabolism , Animals , Blood Vessels/metabolism , Brain/metabolism , Heart Diseases/metabolism , Humans , Inflammation/metabolism , Kidney/metabolism , Kidney Diseases/metabolism
20.
Hypertension ; 71(6): 1075-1082, 2018 06.
Article in English | MEDLINE | ID: mdl-29661841

ABSTRACT

The activity of the renin-angiotensin-aldosterone system is triggered by the release of the protease renin from the kidneys, which in turn is controlled in the sense of negative feedback loops. It is widely assumed that Ang II (angiotensin II) directly inhibits renin expression and secretion via a short-loop feedback by an effect on renin-producing cells (RPCs) mediated by AT1 (Ang II type 1) receptors. Because the concept of such a direct short-loop negative feedback control, which originates mostly from in vitro experiments, has not yet been systematically proven in vivo, we aimed to test the validity of this concept by studying the regulation of renin synthesis and secretion in mice lacking Ang II-AT1 receptors on RPCs. We found that RPCs of the kidney express Ang II-AT1 receptors. Mice with conditional deletion of Ang II-AT1 receptors in RPCs were normal with regard to the number of renin cells, renal renin mRNA, and plasma renin concentrations. Renin expression and secretion of these mice responded to Ang I (angiotensin I)-converting enzyme inhibition and to Ang II infusion like in wild-type (WT) controls. In summary, we did not obtain evidence that Ang II-AT1 receptors on RPCs are of major relevance for the normal regulation of renin expression and secretion in mice. Therefore, we doubt the existence of a direct negative feedback function of Ang II on RPCs.


Subject(s)
Angiotensin II/pharmacology , Angiotensin-Converting Enzyme Inhibitors/pharmacology , Blood Pressure/physiology , Hypertension/metabolism , Receptors, Angiotensin/metabolism , Renin-Angiotensin System/physiology , Renin/blood , Animals , Disease Models, Animal , Female , Hypertension/drug therapy , Hypertension/physiopathology , Immunohistochemistry , Male , Mice , Renin-Angiotensin System/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...