Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 72
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Kidney Int ; 104(6): 1135-1149, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-37843477

RESUMEN

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.


Asunto(s)
Diabetes Mellitus Tipo 2 , Nefropatías Diabéticas , Insuficiencia Renal , Humanos , Animales , Ratones , Nefropatías Diabéticas/etiología , Ácido Láctico , Albuminuria/etiología , Antagonistas de Receptores de Angiotensina/farmacología , Antagonistas de Receptores de Angiotensina/uso terapéutico , Diabetes Mellitus Tipo 2/complicaciones , Inhibidores de la Enzima Convertidora de Angiotensina/farmacología , Inhibidores de la Enzima Convertidora de Angiotensina/uso terapéutico , Riñón , Isoformas de Proteínas
2.
Curr Hypertens Rep ; 25(9): 251-261, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37303020

RESUMEN

PURPOSE OF REVIEW: We summarized recent available data to assess the association between assisted reproductive technology (ART) and risk for preeclampsia. RECENT FINDINGS: The majority of clinical studies supporting the association of preeclampsia and ART are retrospective. Published data from both clinical and pre-clinical studies suggest specific ART procedures may contribute to the increased risk, including in vitro embryo handling and development, hormone stimulation, transfer cycle types, and use of donor oocytes/embryos. Potential mechanisms include epigenetic aberrations leading to abnormal placentation, absence of factors secreted by the corpus luteum, and immunologic responses to allogenic gametes. There is an increased risk of preeclampsia following ART. Treatment plans that favor reduced preeclampsia risk should be considered for ART pregnancies. To make ART pregnancies safer, additional clinical and animal model studies are needed to elucidate the underpinnings of this risk association.


Asunto(s)
Hipertensión , Preeclampsia , Embarazo , Femenino , Humanos , Preeclampsia/etiología , Estudios Retrospectivos , Hipertensión/etiología , Técnicas Reproductivas Asistidas/efectos adversos
3.
Clin Nephrol ; 100(5): 231-234, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37779451

RESUMEN

The COVID-19 pandemic has had lingering effects in healthcare. During the pandemic, inpatient bed shortages became a major barrier to providing care, and access to outpatient dialysis was delayed due to a shortage of staff. This resulted in lengthy admissions to the hospital while awaiting outpatient dialysis placement in a time when hospital bedspace was a scarce resource. In this report we describe the use of the Hospital at Home program for inpatient dialysis care as a novel method to both provide high quality patient care at home and also open inpatient beds in the hospital. Here, we present two cases where this program served both the needs of the patient and the hospital system by providing dialysis care to patients participating in the Hospital at Home program. One was a patient with a recent kidney transplant awaiting improvement in kidney function following transplant. The other was a new mother who was diagnosed with atypical hemolytic uremic syndrome and was awaiting recovery from her acute kidney injury after receiving complement inhibitor therapy. This program has been functional for the past 13 months and has served 37 patients and saved 1,007 inpatient days during that period. Each inpatient day provided by Hospital at Home represents bed space that was made available to another patient in need.


Asunto(s)
COVID-19 , Diálisis Renal , Femenino , Humanos , Diálisis Renal/métodos , Pandemias , COVID-19/epidemiología , Hospitalización , Hospitales
4.
J Am Soc Nephrol ; 33(3): 584-600, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35064051

RESUMEN

BACKGROUND: Mutations in the ubiquitin ligase scaffold protein Cullin 3 (CUL3) gene cause the disease familial hyperkalemic hypertension (FHHt). In the kidney, mutant CUL3 (CUL3-Δ9) increases abundance of With-No-Lysine (K) Kinase 4 (WNK4), inappropriately activating sterile 20/SPS-1-related proline/alanine-rich kinase (SPAK), which then phosphorylates and hyperactivates the Na+Cl- cotransporter (NCC). The precise mechanism by which CUL3-Δ9 causes FHHt is unclear. We tested the hypothesis that reduced abundance of CUL3 and of Kelch-like 3 (KLHL3), the CUL3 substrate adaptor for WNK4, is mechanistically important. Because JAB1, an enzyme that inhibits CUL3 activity by removing the ubiquitin-like protein NEDD8, cannot interact with CUL3-Δ9, we also determined whether Jab1 disruption mimicked the effects of CUL3-Δ9 expression. METHODS: We used an inducible renal tubule-specific system to generate several mouse models expressing CUL3-Δ9, mice heterozygous for both CUL3 and KLHL3 (Cul3+/-/Klhl3+/- ), and mice with short-term Jab1 disruption (to avoid renal injury associated with long-term disruption). RESULTS: Renal KLHL3 was higher in Cul3-/- mice, but lower in Cul3-/-/Δ9 mice and in the Cul3+/-/Δ9 FHHt model, suggesting KLHL3 is a target for both WT and mutant CUL3. Cul3+/-/Klhl3+/- mice displayed increased WNK4-SPAK activation and phospho-NCC abundance and an FHHt-like phenotype with increased plasma [K+] and salt-sensitive blood pressure. Short-term Jab1 disruption in mice lowered the abundance of CUL3 and KLHL3 and increased the abundance of WNK4 and phospho-NCC. CONCLUSIONS: Jab1-/- mice and Cul3+/-/Klhl3+/- mice recapitulated the effects of CUL3-Δ9 expression on WNK4-SPAK-NCC. Our data suggest degradation of both KLHL3 and CUL3 plays a central mechanistic role in CUL3-Δ9-mediated FHHt.


Asunto(s)
Proteínas Cullin , Hipertensión , Seudohipoaldosteronismo , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Proteínas Cullin/genética , Proteínas Cullin/metabolismo , Femenino , Humanos , Hipertensión/genética , Masculino , Ratones , Proteínas de Microfilamentos/genética , Proteínas Serina-Treonina Quinasas/genética , Seudohipoaldosteronismo/genética , Seudohipoaldosteronismo/metabolismo , Miembro 3 de la Familia de Transportadores de Soluto 12/metabolismo
5.
Am J Physiol Renal Physiol ; 323(4): F468-F478, 2022 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-35900342

RESUMEN

The renin-angiotensin-aldosterone and arginine vasopressin-V2 receptor-aquaporin-2 (AQP2) systems converge on the epithelial Na+ channel (ENaC) to regulate blood pressure and plasma tonicity. Although it is established that V2 receptors initiate renal water reabsorption through AQP2, whether V2 receptors can also induce renal Na+ retention through ENaC and raise blood pressure remains an open question. We hypothesized that a specific increase in V2 receptor-mediated ENaC activity can lead to high blood pressure. Our approach was to test effects of chronic activation of V2 receptors in Liddle mice, a genetic mouse model of high ENaC activity, and compare differences in ENaC activity, urine Na+ excretion, and blood pressure with control mice. We found that ENaC activity was elevated in Liddle mice and could not be stimulated further by administration of desmopressin (dDAVP), a V2 receptor-specific agonist. In contrast, Liddle mice showed higher levels of expression of AQP2 and aquaporin-3, but they could still respond to dDAVP infusion by increasing phospho-AQP2 expression. With dDAVP infusion, Liddle mice excreted smaller urine volume and less urine Na+ and developed higher blood pressure compared with control mice; this hypertension was attenuated with administration of the ENaC inhibitor benzamil. We conclude that V2 receptors contribute to hypertension in the Liddle mouse model by promoting primary Na+ and concomitant water retention.NEW & NOTEWORTHY Liddle syndrome is a classic model for hypertension from high epithelial Na+ channel (ENaC) activity. In the Liddle mouse model, vasopressin-2 receptors stimulate both ENaC and aquaporin-2, which increases Na+ and water retention to such an extent that hypertension ensues. Liddle mice will preserve plasma tonicity at the expense of a higher blood pressure; these data highlight the inherent limitation in which the kidney must use ENaC as a pathway to regulate both plasma tonicity and blood pressure.


Asunto(s)
Hipertensión , Desequilibrio Hidroelectrolítico , Animales , Acuaporina 2 , Desamino Arginina Vasopresina/farmacología , Canales Epiteliales de Sodio/metabolismo , Ratones , Receptores de Vasopresinas/metabolismo , Sodio/metabolismo , Agua/metabolismo
6.
Am J Physiol Renal Physiol ; 321(1): F69-F81, 2021 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-34056928

RESUMEN

The renal nephron consists of a series of distinct cell types that function in concert to maintain fluid and electrolyte balance and blood pressure. The renin-angiotensin system (RAS) is central to Na+ and volume balance. We aimed to determine how loss of angiotensin II signaling in the proximal tubule (PT), which reabsorbs the bulk of filtered Na+ and volume, impacts solute transport throughout the nephron. We hypothesized that PT renin-angiotensin system disruption would not only depress PT Na+ transporters but also impact downstream Na+ transporters. Using a mouse model in which the angiotensin type 1a receptor (AT1aR) is deleted specifically within the PT (AT1aR PTKO), we profiled the abundance of Na+ transporters, channels, and claudins along the nephron. Absence of PT AT1aR signaling was associated with lower abundance of PT transporters (Na+/H+ exchanger isoform 3, electrogenic Na+-bicarbonate cotransporter 1, and claudin 2) as well as lower abundance of downstream transporters (total and phosphorylated Na+-K+-2Cl- cotransporter, medullary Na+-K+-ATPase, phosphorylated NaCl cotransporter, and claudin 7) versus controls. However, transport activities of Na+-K+-2Cl- cotransporter and NaCl cotransporter (assessed with diuretics) were similar between groups in order to maintain electrolyte balance. Together, these results demonstrate the primary impact of angiotensin II regulation on Na+ reabsorption in the PT at baseline and the associated influence on downstream Na+ transporters, highlighting the ability of the nephron to integrate Na+ transport along the nephron to maintain homeostasis.NEW & NOTEWORTHY Our study defines a novel role for proximal tubule angiotensin receptors in regulating the abundance of Na+ transporters throughout the nephron, thereby contributing to the integrated control of fluid balance in vivo.


Asunto(s)
Angiotensina II/farmacología , Proteínas de Transporte de Membrana/metabolismo , Nefronas/metabolismo , Miembro 3 de la Familia de Transportadores de Soluto 12/metabolismo , Animales , Riñón/metabolismo , Natriuresis/efectos de los fármacos , Intercambiadores de Sodio-Hidrógeno/metabolismo
7.
Am J Physiol Renal Physiol ; 320(6): F1080-F1092, 2021 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-33969697

RESUMEN

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.


Asunto(s)
Angiotensina II/farmacología , Células Epiteliales/metabolismo , Riñón/irrigación sanguínea , Miocitos del Músculo Liso/metabolismo , Receptor de Angiotensina Tipo 1/metabolismo , Amilorida/farmacología , Animales , Bloqueadores del Canal de Sodio Epitelial/farmacología , Femenino , Furosemida/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Proteínas Fluorescentes Verdes , Hipertensión/inducido químicamente , Proteínas Luminiscentes , Masculino , Ratones , Ratones Endogámicos , Ratones Noqueados , Receptor de Angiotensina Tipo 1/genética , Sodio/metabolismo , Inhibidores del Simportador de Cloruro Sódico y Cloruro Potásico/farmacología , Proteína Fluorescente Roja
8.
J Am Soc Nephrol ; 31(4): 783-798, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32086277

RESUMEN

BACKGROUND: Increased nerve activity causes hypertension and kidney disease. Recent studies suggest that renal denervation reduces BP in patients with hypertension. Renal NE release is regulated by prejunctional α2A-adrenoceptors on sympathetic nerves, and α2A-adrenoceptors act as autoreceptors by binding endogenous NE to inhibit its own release. However, the role of α2A-adrenoceptors in the pathogenesis of hypertensive kidney disease is unknown. METHODS: We investigated effects of α2A-adrenoceptor-regulated renal NE release on the development of angiotensin II-dependent hypertension and kidney disease. In uninephrectomized wild-type and α2A-adrenoceptor-knockout mice, we induced hypertensive kidney disease by infusing AngII for 28 days. RESULTS: Urinary NE excretion and BP did not differ between normotensive α2A-adrenoceptor-knockout mice and wild-type mice at baseline. However, NE excretion increased during AngII treatment, with the knockout mice displaying NE levels that were significantly higher than those of wild-type mice. Accordingly, the α2A-adrenoceptor-knockout mice exhibited a systolic BP increase, which was about 40 mm Hg higher than that found in wild-type mice, and more extensive kidney damage. In isolated kidneys, AngII-enhanced renal nerve stimulation induced NE release and pressor responses to a greater extent in kidneys from α2A-adrenoceptor-knockout mice. Activation of specific sodium transporters accompanied the exaggerated hypertensive BP response in α2A-adrenoceptor-deficient kidneys. These effects depend on renal nerves, as demonstrated by reduced severity of AngII-mediated hypertension and improved kidney function observed in α2A-adrenoceptor-knockout mice after renal denervation. CONCLUSIONS: Our findings reveal a protective role of prejunctional inhibitory α2A-adrenoceptors in pathophysiologic conditions with an activated renin-angiotensin system, such as hypertensive kidney disease, and support the concept of sympatholytic therapy as a treatment.


Asunto(s)
Hipertensión Renal/etiología , Hipertensión Renal/prevención & control , Nefritis/etiología , Nefritis/prevención & control , Receptores Adrenérgicos alfa 2/fisiología , Sistema Nervioso Simpático/fisiopatología , Transmisión Sináptica/fisiología , Angiotensina II , Animales , Modelos Animales de Enfermedad , Hipertensión Renal/fisiopatología , Ratones , Ratones Noqueados , Nefritis/fisiopatología , Simpatectomía
9.
J Am Soc Nephrol ; 31(7): 1479-1495, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32540856

RESUMEN

BACKGROUND: Genetic mutations in α-actinin-4 (ACTN4)-an important actin crosslinking cytoskeletal protein that provides structural support for kidney podocytes-have been linked to proteinuric glomerulosclerosis in humans. However, the effect of post-translational modifications of ACTN4 on podocyte integrity and kidney function is not known. METHODS: Using mass spectrometry, we found that ACTN4 is phosphorylated at serine (S) 159 in human podocytes. We used phosphomimetic and nonphosphorylatable ACTN4 to comprehensively study the effects of this phosphorylation in vitro and in vivo. We conducted x-ray crystallography, F-actin binding and bundling assays, and immunofluorescence staining to evaluate F-actin alignment. Microfluidic organ-on-a-chip technology was used to assess for detachment of podocytes simultaneously exposed to fluid flow and cyclic strain. We then used CRISPR/Cas9 to generate mouse models and assessed for renal injury by measuring albuminuria and examining kidney histology. We also performed targeted mass spectrometry to determine whether high extracellular glucose or TGF-ß levels increase phosphorylation of ACTN4. RESULTS: Compared with the wild type ACTN4, phosphomimetic ACTN4 demonstrated increased binding and bundling activity with F-actin in vitro. Phosphomimetic Actn4 mouse podocytes exhibited more spatially correlated F-actin alignment and a higher rate of detachment under mechanical stress. Phosphomimetic Actn4 mice developed proteinuria and glomerulosclerosis after subtotal nephrectomy. Moreover, we found that exposure to high extracellular glucose or TGF-ß stimulates phosphorylation of ACTN4 at S159 in podocytes. CONCLUSIONS: These findings suggest that increased phosphorylation of ACTN4 at S159 leads to biochemical, cellular, and renal pathology that is similar to pathology resulting from human disease-causing mutations in ACTN4. ACTN4 may mediate podocyte injury as a consequence of both genetic mutations and signaling events that modulate phosphorylation.


Asunto(s)
Actinina/metabolismo , Albuminuria/metabolismo , Glomeruloesclerosis Focal y Segmentaria/metabolismo , Podocitos/metabolismo , Procesamiento Proteico-Postraduccional , Actinina/genética , Actinas/metabolismo , Actinas/ultraestructura , Albuminuria/etiología , Albuminuria/patología , Animales , Células Cultivadas , Femenino , Glomeruloesclerosis Focal y Segmentaria/etiología , Glomeruloesclerosis Focal y Segmentaria/patología , Glucosa/farmacología , Humanos , Dispositivos Laboratorio en un Chip , Masculino , Ratones , Nefrectomía/efectos adversos , Peptidomiméticos , Fosforilación/efectos de los fármacos , Unión Proteica , Serina/metabolismo , Factor de Crecimiento Transformador beta/farmacología
10.
Am J Physiol Renal Physiol ; 314(4): F531-F542, 2018 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-29187372

RESUMEN

Diabetic nephropathy is a major cause of end-stage renal disease in developed countries. While angiotensin-converting enzyme (ACE) inhibitors are used to treat diabetic nephropathy, how intrarenal ACE contributes to diabetic renal injury is uncertain. Here, two mouse models with different patterns of renal ACE expression were studied to determine the specific contribution of tubular vs. glomerular ACE to early diabetic nephropathy: it-ACE mice, which make endothelial ACE but lack ACE expression by renal tubular epithelium, and ACE 3/9 mice, which lack endothelial ACE and only express renal ACE in tubular epithelial cells. The absence of endothelial ACE normalized the glomerular filtration rate and endothelial injury in diabetic ACE 3/9 mice. However, these mice developed tubular injury and albuminuria and displayed low renal levels of megalin that were similar to those observed in diabetic wild-type mice. In diabetic it-ACE mice, despite hyperfiltration, the absence of renal tubular ACE greatly reduced tubulointerstitial injury and albuminuria and increased renal megalin expression compared with diabetic wild-type and diabetic ACE 3/9 mice. These findings demonstrate that endothelial ACE is a central regulator of the glomerular filtration rate while tubular ACE is a key player in the development of tubular injury and albuminuria. These data suggest that tubular injury, rather than hyperfiltration, is the main cause of microalbuminuria in early diabetic nephropathy.


Asunto(s)
Albuminuria/enzimología , Diabetes Mellitus Experimental/enzimología , Nefropatías Diabéticas/enzimología , Túbulos Renales/enzimología , Peptidil-Dipeptidasa A/metabolismo , Albuminuria/genética , Albuminuria/patología , Albuminuria/fisiopatología , Animales , Diabetes Mellitus Experimental/inducido químicamente , Diabetes Mellitus Experimental/genética , Nefropatías Diabéticas/genética , Nefropatías Diabéticas/patología , Nefropatías Diabéticas/fisiopatología , Células Endoteliales/enzimología , Tasa de Filtración Glomerular , Glomérulos Renales/enzimología , Glomérulos Renales/fisiopatología , Túbulos Renales/patología , Túbulos Renales/fisiopatología , Proteína 2 Relacionada con Receptor de Lipoproteína de Baja Densidad/metabolismo , Ratones Noqueados , Peptidil-Dipeptidasa A/deficiencia , Peptidil-Dipeptidasa A/genética , ARN Interferente Pequeño/genética , Estreptozocina
11.
Curr Opin Nephrol Hypertens ; 27(1): 1-7, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29045337

RESUMEN

PURPOSE OF REVIEW: The renin-angiotensin system (RAS) plays a critical role in the pathogenesis of hypertension. Homeostatic actions of the RAS, such as increasing blood pressure (BP) and vasoconstriction, are mediated via type 1 (AT1) receptors for angiotensin II. All components of the RAS are present in the renal proximal tubule, which reabsorbs the bulk of the glomerular filtrate, making this segment of the nephron a location of great interest for solute handling under RAS influence. This review highlights recent studies that illustrate the key role of renal proximal tubule AT1 receptors in BP regulation. RECENT FINDINGS: A variety of investigative approaches have demonstrated that angiotensin II signaling via AT1a receptors, specifically in the renal proximal tubule, is a major regulator of BP and sodium homeostasis. Reduction of proximal tubule AT1a receptors led to lower BPs, whereas overexpression generally caused increased BPs. SUMMARY: AT1a receptors in the proximal tubule are critical to the regulation of BP by the kidney and the RAS. The pattern of BP modulation is associated with alterations in sodium transporters. As a key site for sodium homeostasis, the renal proximal tubule could hence be a potential target in the treatment of hypertension.


Asunto(s)
Presión Sanguínea , Túbulos Renales Proximales/metabolismo , Receptor de Angiotensina Tipo 1/metabolismo , Sistema Renina-Angiotensina/fisiología , Animales , Homeostasis , Humanos , Receptor de Angiotensina Tipo 1/fisiología , Transducción de Señal , Sodio/metabolismo
12.
FASEB J ; 31(12): 5520-5529, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28842425

RESUMEN

Accurate analysis of placental and fetal oxygenation is critical during pregnancy. Photoacoustic imaging (PAI) combines laser technology with ultrasound in real time. We tested the sensitivity and accuracy of PAI for analysis of placental and fetal oxygen saturation (sO2) in mice. The placental labyrinth (L) had a higher sO2 than the junctional zone plus decidua region (JZ+D) in C57Bl/6 mice. Changing maternal O2 from 100 to 20% in C57Bl/6 mice lowered sO2 in these regions. C57Bl/6 mice were treated with the NO synthase inhibitor L-NG-nitroarginine methyl ester (L-NAME) from gestational day (GD) 11 to GD18 to induce hypertension. L-NAME decreased sO2 in L and JZ+D at GD14 and GD18 in association with fetal growth restriction and higher blood pressure. Hypoxia-inducible factor 1α immunostaining was higher in L-NAME vs control mice at GD14. Fetal sO2 levels were similar between l-NAME and control mice at GD14 and GD18. In contrast to untreated C57Bl/6, L-NAME decreased placental sO2 at GD14 and GD18 vs GD10 or GD12. Placental sO2 was lower in fetal growth restriction in an angiotensin-converting enzyme 2 knockout mouse model characterized by placental hypoxia. On phantom studies, patterns of sO2 measured directly correlated with those measured by PAI. In summary, PAI enables the detection of placental and fetal oxygenation during normal and pathologic pregnancies in mice.-Yamaleyeva, L. M., Sun, Y., Bledsoe, T., Hoke, A., Gurley, S. B., Brosnihan, K. B. Photoacoustic imaging for in vivo quantification of placental oxygenation in mice.


Asunto(s)
Oxígeno/metabolismo , Técnicas Fotoacústicas/métodos , Placenta/metabolismo , Animales , Modelos Animales de Enfermedad , Femenino , Retardo del Crecimiento Fetal/metabolismo , Hipertensión/inducido químicamente , Hipertensión/metabolismo , Hipoxia/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , NG-Nitroarginina Metil Éster/farmacología , Placenta/efectos de los fármacos , Embarazo
13.
J Exp Ther Oncol ; 11(2): 107-115, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28976133

RESUMEN

OBJECTIVE: The renin-angiotensin system, through its type 1 and type 2 angiotensin receptors (AT1R and AT2R, respectively) may have a role in prostate cancer. The objective of this pilot study was to explore that potential role by determining whether the AT1R blocker, losartan, would reduce the growth of LAPC-4 prostate cancer xenografts in nude mice. We also evaluated the tumor growth effects of using angiotensin II to activate both AT1R and AT2R simultaneously. Our data showed that losartan decreased tumor volumes by 56% versus control. This decrease reached statistical significance at day 54 (p = 0.0014). By day 54, Ki67 was also reduced in the losartan group, though not significantly so (p = 0.077). Losartan had no significant effect on AT1R or AT2R expression. Despite significant increases in both AT1R and AT2R at day 29 (p = 0.043 and 0.038, respectively), the administration of angiotensin II did not result in any significant differences in tumor volumes or ki67 at any time point. These data suggest that selective activation and induction of AT2R coupled with blockade of AT1R may slow prostate cancer growth. Future larger studies are needed to confirm these results.


Asunto(s)
Bloqueadores del Receptor Tipo 1 de Angiotensina II/farmacología , Losartán/farmacología , Neoplasias de la Próstata/patología , Receptor de Angiotensina Tipo 1/genética , Receptor de Angiotensina Tipo 2/genética , Carga Tumoral/efectos de los fármacos , Angiotensina II/farmacología , Animales , Línea Celular Tumoral , Masculino , Ratones , Trasplante de Neoplasias , Neoplasias de la Próstata/metabolismo , ARN Mensajero/efectos de los fármacos , ARN Mensajero/metabolismo , Receptor de Angiotensina Tipo 1/efectos de los fármacos , Receptor de Angiotensina Tipo 1/metabolismo , Receptor de Angiotensina Tipo 2/efectos de los fármacos , Transducción de Señal , Transcriptoma/efectos de los fármacos , Vasoconstrictores/farmacología
14.
J Am Soc Nephrol ; 27(8): 2257-64, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-26744488

RESUMEN

Inappropriate activation of the renin-angiotensin system (RAS) contributes to many CKDs. However, the role of the RAS in modulating AKI requires elucidation, particularly because stimulating type 1 angiotensin II (AT1) receptors in the kidney or circulating inflammatory cells can have opposing effects on the generation of inflammatory mediators that underpin the pathogenesis of AKI. For example, TNF-α is a fundamental driver of cisplatin nephrotoxicity, and generation of TNF-α is suppressed or enhanced by AT1 receptor signaling in T lymphocytes or the distal nephron, respectively. In this study, cell tracking experiments with CD4-Cre mT/mG reporter mice revealed robust infiltration of T lymphocytes into the kidney after cisplatin injection. Notably, knockout of AT1 receptors on T lymphocytes exacerbated the severity of cisplatin-induced AKI and enhanced the cisplatin-induced increase in TNF-α levels locally within the kidney and in the systemic circulation. In contrast, knockout of AT1 receptors on kidney epithelial cells ameliorated the severity of AKI and suppressed local and systemic TNF-α production induced by cisplatin. Finally, disrupting TNF-α production specifically within the renal tubular epithelium attenuated the AKI and the increase in circulating TNF-α levels induced by cisplatin. These results illustrate discrepant tissue-specific effects of RAS stimulation on cisplatin nephrotoxicity and raise the concern that inflammatory mediators produced by renal parenchymal cells may influence the function of remote organs by altering systemic cytokine levels. Our findings suggest selective inhibition of AT1 receptors within the nephron as a promising intervention for protecting patients from cisplatin-induced nephrotoxicity.


Asunto(s)
Lesión Renal Aguda/fisiopatología , Riñón/metabolismo , Receptor de Angiotensina Tipo 1/fisiología , Linfocitos T , Lesión Renal Aguda/inducido químicamente , Animales , Cisplatino/administración & dosificación , Epitelio/metabolismo , Femenino , Ratones , Receptor de Angiotensina Tipo 1/biosíntesis , Linfocitos T/metabolismo
15.
Curr Opin Nephrol Hypertens ; 25(1): 59-66, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26575394

RESUMEN

PURPOSE OF REVIEW: Angiotensin-converting enzyme 2 (ACE2) is a monocarboxypeptidase that metabolizes angiotensin II (AngII). AngII levels can be modulated by ACE2 in tissues where the enzyme is highly expressed, such as the kidney. In the kidney, ACE2 has the potential to regulate the intrarenal renin-angiotensin system (RAS), which can impact blood pressure and renal injury. Thus, in disease states where the RAS is often upregulated, the function of ACE2 plays a particularly important role. This review highlights the results of recent studies that demonstrate the interplay between ACE2 and the kidney under normal and pathological conditions. RECENT FINDINGS: ACE2 has been reported to play a key role in renal and cardiovascular function. Recent studies have implicated shedding of the membrane-bound ectodomain of ACE2 as one way in which the enzyme can be regulated and enzymatic activity altered. This posttranslational modification releases a fragment which retains enzymatic activity, and thus provides a novel mechanism by which the RAS can be altered in response to physiological stimuli. Decreased ACE2 activity is associated with increased blood pressure, diabetes, and oxidative stress, whereas, increased levels of ACE2 appear to be renoprotective. SUMMARY: A growing body of evidence, involving both experimental and human studies, points out the crucial role that ACE2 plays on the modulation of renal injury. Thus, therapeutic targets aiming to increase ACE2 activity and the ACE2-Ang(1-7)-MasR axis could potentially become clinically relevant, especially in disease states where the renal RAS is upregulated.


Asunto(s)
Enfermedades Renales/etiología , Peptidil-Dipeptidasa A/fisiología , Enzima Convertidora de Angiotensina 2 , Animales , Presión Sanguínea , Nefropatías Diabéticas/metabolismo , Humanos , Riñón/enzimología , Riñón/metabolismo , Enfermedades Renales/enzimología , Estrés Oxidativo , Peptidil-Dipeptidasa A/química , Peptidil-Dipeptidasa A/deficiencia , Proto-Oncogenes Mas
16.
J Am Soc Nephrol ; 26(12): 2953-62, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25855778

RESUMEN

Inappropriate activation of the type 1A angiotensin (AT1A) receptor contributes to the pathogenesis of hypertension and its associated complications. To define the role for actions of vascular AT1A receptors in BP regulation and hypertension pathogenesis, we generated mice with cell-specific deletion of AT1A receptors in smooth muscle cells (SMKO mice) using Loxp technology and Cre transgenes with robust expression in both conductance and resistance arteries. We found that elimination of AT1A receptors from vascular smooth muscle cells (VSMCs) caused a modest (approximately 7 mmHg) yet significant reduction in baseline BP and exaggerated sodium sensitivity in mice. Additionally, the severity of angiotensin II (Ang II)-dependent hypertension was dramatically attenuated in SMKO mice, and this protection against hypertension was associated with enhanced urinary excretion of sodium. Despite the lower BP, acute vasoconstrictor responses to Ang II in the systemic vasculature were largely preserved (approximately 80% of control levels) in SMKO mice because of exaggerated activity of the sympathetic nervous system rather than residual actions of AT1B receptors. In contrast, Ang II-dependent responses in the renal circulation were almost completely eliminated in SMKO mice (approximately 5%-10% of control levels). These findings suggest that direct actions of AT1A receptors in VSMCs are essential for regulation of renal blood flow by Ang II and highlight the capacity of Ang II-dependent vascular responses in the kidney to effect natriuresis and BP control.


Asunto(s)
Presión Sanguínea/fisiología , Natriuresis/fisiología , Receptor de Angiotensina Tipo 1/fisiología , Circulación Renal/fisiología , Antagonistas Adrenérgicos alfa/farmacología , Angiotensina II/farmacología , Animales , Presión Sanguínea/efectos de los fármacos , Catecolaminas/orina , Hipertensión/inducido químicamente , Hipertensión/fisiopatología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/fisiología , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/fisiología , Natriuresis/efectos de los fármacos , Fentolamina/farmacología , Receptor de Angiotensina Tipo 1/genética , Circulación Renal/efectos de los fármacos , Sistema Nervioso Simpático/efectos de los fármacos , Sistema Nervioso Simpático/fisiopatología , Vasoconstricción/efectos de los fármacos , Vasoconstricción/fisiología , Vasoconstrictores/farmacología
17.
Am J Physiol Endocrinol Metab ; 309(1): E84-94, 2015 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-25968580

RESUMEN

Angiotensin-converting enzyme 2 (ACE2) knockout is associated with reduced fetal weight at late gestation; however, whether uteroplacental vascular and/or hemodynamic disturbances underlie this growth-restricted phenotype is unknown. Uterine artery reactivity and flow velocities, umbilical flow velocities, trophoblast invasion, and placental hypoxia were determined in ACE2 knockout (KO) and C57Bl/6 wild-type (WT) mice at day 14 of gestation. Although systolic blood pressure was higher in pregnant ACE2 KO vs. WT mice (102.3 ± 5.1 vs. 85.1 ± 1.9 mmHg, n = 5-6), the magnitude of difference was similar to that observed in nonpregnant ACE2 KO vs. WT mice. Maternal urinary protein excretion, serum creatinine, and kidney or heart weights were not different in ACE2 KO vs. WT. Fetal weight and pup-to-placental weight ratio were lower in ACE2 KO vs. WT mice. A higher sensitivity to Ang II [pD2 8.64 ± 0.04 vs. 8.5 ± 0.03 (-log EC50)] and greater maximal contraction to phenylephrine (169.0 ± 9.0 vs. 139.0 ± 7.0% KMAX), were associated with lower immunostaining for Ang II receptor 2 and fibrinoid content of the uterine artery in ACE2 KO mice. Uterine artery flow velocities and trophoblast invasion were similar between study groups. In contrast, umbilical artery peak systolic velocities (60.2 ± 4.5 vs. 75.1 ± 4.5 mm/s) and the resistance index measured using VEVO 2100 ultrasound were lower in the ACE2 KO vs. WT mice. Immunostaining for pimonidazole, a marker of hypoxia, and hypoxia-inducible factor-2α were higher in the trophospongium and placental labyrinth of the ACE2 KO vs. WT. In summary, placental hypoxia and uterine artery dysfunction develop before major growth of the fetus occurs and may explain the fetal growth restricted phenotype.


Asunto(s)
Hipoxia/genética , Peptidil-Dipeptidasa A/genética , Placenta/patología , Cordón Umbilical/irrigación sanguínea , Arteria Uterina/fisiopatología , Enzima Convertidora de Angiotensina 2 , Animales , Velocidad del Flujo Sanguíneo , Femenino , Retardo del Crecimiento Fetal/genética , Retardo del Crecimiento Fetal/metabolismo , Retardo del Crecimiento Fetal/patología , Retardo del Crecimiento Fetal/fisiopatología , Hipoxia/metabolismo , Hipoxia/fisiopatología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Placenta/irrigación sanguínea , Placenta/metabolismo , Placenta/fisiopatología , Circulación Placentaria/fisiología , Embarazo , Cordón Umbilical/fisiopatología
18.
Arterioscler Thromb Vasc Biol ; 34(12): 2617-23, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25301841

RESUMEN

OBJECTIVE: Angiotensin-converting enzyme 2 (ACE2) cleaves angiotensin II (AngII) to form angiotensin-(1-7) (Ang-(1-7)), which generally opposes effects of AngII. AngII infusion into hypercholesterolemic male mice induces formation of abdominal aortic aneurysms (AAAs). This study tests the hypothesis that deficiency of ACE2 promotes AngII-induced AAAs, whereas ACE2 activation suppresses aneurysm formation. APPROACH AND RESULTS: ACE2 protein was detectable by immunostaining in mice and human AAAs. Whole-body deficiency of ACE2 significantly increased aortic lumen diameters and external diameters of suprarenal aortas from AngII-infused mice. Conversely, ACE2 deficiency in bone marrow-derived cells had no effect on AngII-induced AAAs. In contrast to AngII-induced AAAs, ACE2 deficiency had no significant effect on external aortic diameters of elastase-induced AAAs. Because ACE2 deficiency promoted AAA formation in AngII-infused mice, we determined whether ACE2 activation suppressed AAAs. ACE2 activation by administration of diminazene aceturate (30 mg/kg per day) to Ldlr(-/-) mice increased kidney ACE2 mRNA abundance and activity and elevated plasma Ang-(1-7) concentrations. Unexpectedly, administration of diminazene aceturate significantly reduced total sera cholesterol and very low-density lipoprotein-cholesterol concentrations. Notably, diminazene aceturate significantly decreased aortic lumen diameters and aortic external diameters of AngII-infused mice resulting in a marked reduction in AAA incidence (from 73% to 29%). None of these effects of diminazene aceturate were observed in the Ace2(-/y) mice. CONCLUSIONS: These results demonstrate that ACE2 exerts a modulatory role in AngII-induced AAA formation, and that therapeutic stimulation of ACE2 could be a benefit to reduce AAA expansion and rupture in patients with an activated renin-angiotensin system.


Asunto(s)
Aneurisma de la Aorta Abdominal/etiología , Aneurisma de la Aorta Abdominal/metabolismo , Peptidil-Dipeptidasa A/metabolismo , Anciano , Angiotensina II/metabolismo , Enzima Convertidora de Angiotensina 2 , Animales , Aneurisma de la Aorta Abdominal/patología , Diminazeno/análogos & derivados , Diminazeno/farmacología , Modelos Animales de Enfermedad , Activación Enzimática/efectos de los fármacos , Femenino , Humanos , Hipercolesterolemia/complicaciones , Hipercolesterolemia/metabolismo , Leucocitos/enzimología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , Peptidil-Dipeptidasa A/deficiencia , Peptidil-Dipeptidasa A/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores de LDL/deficiencia , Receptores de LDL/genética , Sistema Renina-Angiotensina
19.
Neurourol Urodyn ; 34(1): 72-8, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25646557

RESUMEN

AIMS: Diabetes is associated with both dysfunction of the lower urinary tract (LUT) and overactivity of the renin-angiotensin system (RAS). Although it is well known that the RAS affects normal LUT function, very little is known about RAS effects on the diabetic LUT. Accordingly, we investigated the effects of chronic angiotensin II (AngII) treatment on the LUT in a model of type 1 diabetes. METHODS: Ins2(Akita) diabetic mice (20 weeks old) and their age-matched background controls underwent conscious cystometric evaluation after 4 weeks of chronic AngII treatment (700 ng/kg/min by osmotic pump) or vehicle (saline). RESULTS: Diabetic mice had compensated LUT function with bladder hypertrophy. Specifically, micturition volume, residual volume, and bladder capacity were all increased, while voiding efficiency and pressure generation were unchanged as bladder mass, contraction duration, and phasic urethral function were increased. AngII significantly increased voiding efficiency and peak voiding pressure and decreased phasic frequency irrespective of diabetic state and, in diabetic but not normoglycemic control mice, significantly decreased residual volume and increased contraction duration and nonphasic contraction duration. CONCLUSIONS: The Ins2(Akita) diabetic mice had compensated LUT function at 20 weeks of age. Even under these conditions, AngII had beneficial effects on LUT function, resulting in increased voiding efficiency. Future studies should therefore be conducted to determine whether AngII can rescue the decompensated LUT function occurring in end-stage diabetic uropathy.


Asunto(s)
Angiotensina II/administración & dosificación , Diabetes Mellitus Experimental/fisiopatología , Diabetes Mellitus Tipo 1/fisiopatología , Vejiga Urinaria/efectos de los fármacos , Vejiga Urinaria/fisiopatología , Micción/efectos de los fármacos , Animales , Masculino , Ratones , Enfermedades de la Vejiga Urinaria/fisiopatología , Micción/fisiología
20.
Hypertension ; 81(4): 682-686, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38507510

RESUMEN

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.


Asunto(s)
Nefropatías Diabéticas , Hipertensión , Humanos , Sistema Renina-Angiotensina , Promoción de la Salud , Riñón/metabolismo , Renina/metabolismo , Nefropatías Diabéticas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA