Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
1.
J Pharmacol Exp Ther ; 2024 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-38849142

RESUMEN

Acute Kidney Injury (AKI) is characterized by an abrupt decline in kidney function and has been associated with excess risks of death, kidney disease progression, and cardiovascular events. The kidney has a high energetic demand with mitochondrial health being essential to renal function and damaged mitochondria has been reported across AKI subtypes. 5' adenosine monophosphate-activated protein kinase (AMPK) activation preserves cellular energetics through improvement of mitochondrial function and biogenesis when ATP levels are low such as under ischemia-induced AKI. We developed a selective potent small molecule pan AMPK activator, compound 1, and tested its ability to increase AMPK activity and preserve kidney function during ischemia/reperfusion injury in rats. A single administration of 1 caused sustained activation of AMPK for at least 24 hours, protected against acute tubular necrosis, and reduced clinical markers of tubular injury such as NephroCheck and Fractional Excretion of Sodium (FENa). Reduction in plasma creatinine and increased Glomerular Filtration Rate (GFR) indicated preservation of kidney function. Surprisingly, we observed a strong diuretic effect of AMPK activation associated with natriuresis both with and without AKI. Our findings demonstrate that activation of AMPK leads to protection of tubular function under hypoxic/ischemic conditions which holds promise as a potential novel therapeutic approach for AKI. Significance Statement No approved pharmacological therapies currently exist for acute kidney injury. We developed Compound 1 which dose-dependently activated AMPK in the kidney and protected kidney function and tubules after ischemic renal injury in the rat. This was accompanied by natriuresis in injured as well as uninjured rats.

2.
J Am Chem Soc ; 145(38): 20825-20836, 2023 09 27.
Artículo en Inglés | MEDLINE | ID: mdl-37589185

RESUMEN

During fibroproliferation, protein-associated extracellular aldehydes are formed by the oxidation of lysine residues on extracellular matrix proteins to form the aldehyde allysine. Here we report three Mn(II)-based, small-molecule magnetic resonance probes that contain α-effect nucleophiles to target allysine in vivo and report on tissue fibrogenesis. We used a rational design approach to develop turn-on probes with a 4-fold increase in relaxivity upon targeting. The effects of aldehyde condensation rate and hydrolysis kinetics on the performance of the probes to detect tissue fibrogenesis non-invasively in mouse models were evaluated by a systemic aldehyde tracking approach. We showed that, for highly reversible ligations, off-rate was a stronger predictor of in vivo efficiency, enabling histologically validated, three-dimensional characterization of pulmonary fibrogenesis throughout the entire lung. The exclusive renal elimination of these probes allowed for rapid imaging of liver fibrosis. Reducing the hydrolysis rate by forming an oxime bond with allysine enabled delayed phase imaging of kidney fibrogenesis. The imaging efficacy of these probes, coupled with their rapid and complete elimination from the body, makes them strong candidates for clinical translation.


Asunto(s)
Ácido 2-Aminoadípico , Aldehídos , Ratones , Animales , Ácido 2-Aminoadípico/química , Imagen por Resonancia Magnética , Pulmón
3.
J Biol Chem ; 295(10): 3115-3133, 2020 03 06.
Artículo en Inglés | MEDLINE | ID: mdl-32005658

RESUMEN

The fortuitously discovered antiaging membrane protein αKlotho (Klotho) is highly expressed in the kidney, and deletion of the Klotho gene in mice causes a phenotype strikingly similar to that of chronic kidney disease (CKD). Klotho functions as a co-receptor for fibroblast growth factor 23 (FGF23) signaling, whereas its shed extracellular domain, soluble Klotho (sKlotho), carrying glycosidase activity, is a humoral factor that regulates renal health. Low sKlotho in CKD is associated with disease progression, and sKlotho supplementation has emerged as a potential therapeutic strategy for managing CKD. Here, we explored the structure-function relationship and post-translational modifications of sKlotho variants to guide the future design of sKlotho-based therapeutics. Chinese hamster ovary (CHO)- and human embryonic kidney (HEK)-derived WT sKlotho proteins had varied activities in FGF23 co-receptor and ß-glucuronidase assays in vitro and distinct properties in vivo Sialidase treatment of heavily sialylated CHO-sKlotho increased its co-receptor activity 3-fold, yet it remained less active than hyposialylated HEK-sKlotho. MS and glycopeptide-mapping analyses revealed that HEK-sKlotho is uniquely modified with an unusual N-glycan structure consisting of N,N'-di-N-acetyllactose diamine at multiple N-linked sites, one of which at Asn-126 was adjacent to a putative GalNAc transfer motif. Site-directed mutagenesis and structural modeling analyses directly implicated N-glycans in Klotho's protein folding and function. Moreover, the introduction of two catalytic glutamate residues conserved across glycosidases into sKlotho enhanced its glucuronidase activity but decreased its FGF23 co-receptor activity, suggesting that these two functions might be structurally divergent. These findings open up opportunities for rational engineering of pharmacologically enhanced sKlotho therapeutics for managing kidney disease.


Asunto(s)
Glucuronidasa/metabolismo , Insuficiencia Renal Crónica/patología , Animales , Células CHO , Dominio Catalítico , Cromatografía Líquida de Alta Presión , Cricetinae , Cricetulus , Factor-23 de Crecimiento de Fibroblastos , Tasa de Filtración Glomerular/efectos de los fármacos , Glucuronidasa/química , Glucuronidasa/genética , Glicopéptidos/análisis , Células HEK293 , Semivida , Humanos , Proteínas Klotho , Espectrometría de Masas , Mutagénesis Sitio-Dirigida , Procesamiento Proteico-Postraduccional , Ratas , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/farmacología , Proteínas Recombinantes/uso terapéutico , Insuficiencia Renal Crónica/metabolismo , Daño por Reperfusión/tratamiento farmacológico , Daño por Reperfusión/patología , Daño por Reperfusión/veterinaria , Relación Estructura-Actividad
4.
Blood ; 130(3): 328-339, 2017 07 20.
Artículo en Inglés | MEDLINE | ID: mdl-28515091

RESUMEN

Angiotensin-converting enzyme (ACE) inhibitors are widely used to reduce blood pressure. Here, we examined if an ACE is important for the antibacterial effectiveness of neutrophils. ACE knockout mice or mice treated with an ACE inhibitor were more susceptible to bacterial infection by methicillin-resistant Staphylococcus aureus (MRSA). In contrast, mice overexpressing ACE in neutrophils (NeuACE mice) have increased resistance to MRSA and better in vitro killing of MRSA, Pseudomonas aeruginosa, and Klebsiella pneumoniae ACE overexpression increased neutrophil production of reactive oxygen species (ROS) following MRSA challenge, an effect independent of the angiotensin II AT1 receptor. Specifically, as compared with wild-type (WT) mice, there was a marked increase of superoxide generation (>twofold, P < .0005) in NeuACE neutrophils following infection, whereas ACE knockout neutrophils decreased superoxide production. Analysis of membrane p47-phox and p67-phox indicates that ACE increases reduced NAD phosphate oxidase activity but does not increase expression of these subunits. Increased ROS generation mediates the enhanced bacterial resistance of NeuACE mice because the enhanced resistance is lost with DPI (an inhibitor of ROS production by flavoenzymes) inhibition. NeuACE granulocytes also have increased neutrophil extracellular trap formation and interleukin-1ß release in response to MRSA. In a mouse model of chemotherapy-induced neutrophil depletion, transfusion of ACE-overexpressing neutrophils was superior to WT neutrophils in treating MRSA infection. These data indicate a previously unknown function of ACE in neutrophil antibacterial defenses and suggest caution in the treatment of certain individuals with ACE inhibitors. ACE overexpression in neutrophils may be useful in boosting the immune response to antibiotic-resistant bacterial infection.


Asunto(s)
Resistencia a la Enfermedad/genética , Inmunidad Innata , Neutrófilos/inmunología , Peptidil-Dipeptidasa A/inmunología , Infecciones Estafilocócicas/inmunología , Superóxidos/inmunología , Animales , Membrana Celular , Trampas Extracelulares/inmunología , Femenino , Regulación de la Expresión Génica , Humanos , Interleucina-1beta/genética , Interleucina-1beta/inmunología , Klebsiella pneumoniae , Masculino , Staphylococcus aureus Resistente a Meticilina/crecimiento & desarrollo , Staphylococcus aureus Resistente a Meticilina/inmunología , Ratones , Ratones Noqueados , NADPH Oxidasas/genética , NADPH Oxidasas/inmunología , Neutrófilos/citología , Neutrófilos/trasplante , Peptidil-Dipeptidasa A/deficiencia , Peptidil-Dipeptidasa A/genética , Fosfoproteínas/genética , Fosfoproteínas/inmunología , Pseudomonas aeruginosa , Receptor de Angiotensina Tipo 1/genética , Receptor de Angiotensina Tipo 1/inmunología , Transducción de Señal , Infecciones Estafilocócicas/enzimología , Infecciones Estafilocócicas/microbiología , Infecciones Estafilocócicas/prevención & control , Superóxidos/metabolismo
5.
J Am Soc Nephrol ; 29(10): 2546-2561, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30185469

RESUMEN

BACKGROUND: Recent evidence emphasizes the critical role of inflammation in the development of diabetic nephropathy. Angiotensin-converting enzyme (ACE) plays an active role in regulating the renal inflammatory response associated with diabetes. Studies have also shown that ACE has roles in inflammation and the immune response that are independent of angiotensin II. ACE's two catalytically independent domains, the N- and C-domains, can process a variety of substrates other than angiotensin I. METHODS: To examine the relative contributions of each ACE domain to the sodium retentive state, renal inflammation, and renal injury associated with diabetic kidney disease, we used streptozotocin to induce diabetes in wild-type mice and in genetic mouse models lacking either a functional ACE N-domain (NKO mice) or C-domain (CKO mice). RESULTS: In response to a saline challenge, diabetic NKO mice excreted 32% more urinary sodium compared with diabetic wild-type or CKO mice. Diabetic NKO mice also exhibited 55% less renal epithelial sodium channel cleavage (a marker of channel activity), 55% less renal IL-1ß, 53% less renal TNF-α, and 53% less albuminuria than diabetic wild-type mice. This protective phenotype was not associated with changes in renal angiotensin II levels. Further, we present evidence that the anti-inflammatory tetrapeptide N-acetyl-seryl-asparyl-lysyl-proline (AcSDKP), an ACE N-domain-specific substrate that accumulates in the urine of NKO mice, mediates the beneficial effects observed in the NKO. CONCLUSIONS: These data indicate that increasing AcSDKP by blocking the ACE N-domain facilitates sodium excretion and ameliorates diabetic kidney disease independent of intrarenal angiotensin II regulation.


Asunto(s)
Nefropatías Diabéticas/metabolismo , Nefropatías Diabéticas/patología , Peptidil-Dipeptidasa A/química , Peptidil-Dipeptidasa A/deficiencia , Sustitución de Aminoácidos , Angiotensina II/metabolismo , Animales , Dominio Catalítico/genética , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patología , Nefropatías Diabéticas/genética , Canales Epiteliales de Sodio/metabolismo , Inflamación/genética , Inflamación/metabolismo , Inflamación/patología , Interleucina-1beta/metabolismo , Riñón/metabolismo , Riñón/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mutagénesis Sitio-Dirigida , Natriuresis/genética , Natriuresis/fisiología , Oligopéptidos/antagonistas & inhibidores , Oligopéptidos/metabolismo , Peptidil-Dipeptidasa A/genética , Dominios Proteicos , Sistema Renina-Angiotensina/fisiología
6.
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
7.
Kidney Int ; 91(4): 856-867, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-27988209

RESUMEN

Renal parenchymal injury predisposes to salt-sensitive hypertension, but how this occurs is not known. Here we tested whether renal tubular angiotensin converting enzyme (ACE), the main site of kidney ACE expression, is central to the development of salt sensitivity in this setting. Two mouse models were used: it-ACE mice in which ACE expression is selectively eliminated from renal tubular epithelial cells; and ACE 3/9 mice, a compound heterozygous mouse model that makes ACE only in renal tubular epithelium from the ACE 9 allele, and in liver hepatocytes from the ACE 3 allele. Salt sensitivity was induced using a post L-NAME salt challenge. While both wild-type and ACE 3/9 mice developed arterial hypertension following three weeks of high salt administration, it-ACE mice remained normotensive with low levels of renal angiotensin II. These mice displayed increased sodium excretion, lower sodium accumulation, and an exaggerated reduction in distal sodium transporters. Thus, in mice with renal injury induced by L-NAME pretreatment, renal tubular epithelial ACE, and not ACE expression by renal endothelium, lung, brain, or plasma, is essential for renal angiotensin II accumulation and salt-sensitive hypertension.


Asunto(s)
Presión Arterial , Hipertensión/enzimología , Túbulos Renales/enzimología , NG-Nitroarginina Metil Éster , Peptidil-Dipeptidasa A/metabolismo , Sistema Renina-Angiotensina , Cloruro de Sodio Dietético , Angiotensina II/metabolismo , Animales , Modelos Animales de Enfermedad , Canales Epiteliales de Sodio/metabolismo , Regulación Enzimológica de la Expresión Génica , Hipertensión/inducido químicamente , Hipertensión/genética , Hipertensión/fisiopatología , Túbulos Renales/fisiopatología , Hígado/enzimología , Ratones Transgénicos , Natriuresis , Peptidil-Dipeptidasa A/deficiencia , Peptidil-Dipeptidasa A/genética , Eliminación Renal , Sistema Renina-Angiotensina/genética , Intercambiador 3 de Sodio-Hidrógeno , Intercambiadores de Sodio-Hidrógeno/metabolismo , Simportadores de Cloruro de Sodio-Potasio/metabolismo , Miembro 3 de la Familia de Transportadores de Soluto 12/metabolismo , Factores de Tiempo
8.
Pharmacol Rev ; 65(1): 1-46, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23257181

RESUMEN

Angiotensin-converting enzyme (ACE) is a zinc-dependent peptidase responsible for converting angiotensin I into the vasoconstrictor angiotensin II. However, ACE is a relatively nonspecific peptidase that is capable of cleaving a wide range of substrates. Because of this, ACE and its peptide substrates and products affect many physiologic processes, including blood pressure control, hematopoiesis, reproduction, renal development, renal function, and the immune response. The defining feature of ACE is that it is composed of two homologous and independently catalytic domains, the result of an ancient gene duplication, and ACE-like genes are widely distributed in nature. The two ACE catalytic domains contribute to the wide substrate diversity of ACE and, by extension, the physiologic impact of the enzyme. Several studies suggest that the two catalytic domains have different biologic functions. Recently, the X-ray crystal structure of ACE has elucidated some of the structural differences between the two ACE domains. This is important now that ACE domain-specific inhibitors have been synthesized and characterized. Once widely available, these reagents will undoubtedly be powerful tools for probing the physiologic actions of each ACE domain. In turn, this knowledge should allow clinicians to envision new therapies for diseases not currently treated with ACE inhibitors.


Asunto(s)
Peptidil-Dipeptidasa A/fisiología , Inhibidores de la Enzima Convertidora de Angiotensina/farmacología , Animales , Historia del Siglo XX , Humanos , Peptidil-Dipeptidasa A/química , Peptidil-Dipeptidasa A/historia , Polimorfismo Genético , Estructura Terciaria de Proteína , Renina/fisiología
9.
J Am Soc Nephrol ; 25(12): 2752-63, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25012170

RESUMEN

The kidney is an important source of angiotensin-converting enzyme (ACE) in many species, including humans. However, the specific effects of local ACE on renal function and, by extension, BP control are not completely understood. We previously showed that mice lacking renal ACE, are resistant to the hypertension induced by angiotensin II infusion. Here, we examined the responses of these mice to the low-systemic angiotensin II hypertensive model of nitric oxide synthesis inhibition with L-NAME. In contrast to wild-type mice, mice without renal ACE did not develop hypertension, had lower renal angiotensin II levels, and enhanced natriuresis in response to L-NAME. During L-NAME treatment, the absence of renal ACE was associated with blunted GFR responses; greater reductions in abundance of proximal tubule Na(+)/H(+) exchanger 3, Na(+)/Pi co-transporter 2, phosphorylated Na(+)/K(+)/Cl(-) cotransporter, and phosphorylated Na(+)/Cl(-) cotransporter; and greater reductions in abundance and processing of the γ isoform of the epithelial Na(+) channel. In summary, the presence of ACE in renal tissue facilitates angiotensin II accumulation, GFR reductions, and changes in the expression levels and post-translational modification of sodium transporters that are obligatory for sodium retention and hypertension in response to nitric oxide synthesis inhibition.


Asunto(s)
Hipertensión/metabolismo , Riñón/metabolismo , Óxido Nítrico Sintasa/antagonistas & inhibidores , Peptidil-Dipeptidasa A/fisiología , Angiotensina II/metabolismo , Animales , Presión Sanguínea , Tasa de Filtración Glomerular , Hipertensión/tratamiento farmacológico , Masculino , Ratones , Ratones Endogámicos C57BL , NG-Nitroarginina Metil Éster/química , Natriuresis , Óxido Nítrico/metabolismo , Fosforilación , Regiones Promotoras Genéticas , Procesamiento Proteico-Postraduccional , Renina/sangre , Simportadores/metabolismo
10.
Am J Respir Cell Mol Biol ; 50(2): 270-80, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24007300

RESUMEN

IL-1ß is a potent proinflammatory cytokine that is implicated in the pathogenesis of acute respiratory distress syndrome. We hypothesized that LPS and mechanical ventilation (MV) together could lead to IL-1ß secretion and the development of acute lung injury (ALI), and that this process would be dependent on caspase-1 and the nucleotide binding domain and leucine-rich repeat (NLR) pyrin domain containing 3 (NLRP3) inflammasome activation. The objectives of this study were to determine the specific role of IL-1ß, caspase-1, and the NLRP3 inflammasome in a two-hit model of ALI due to LPS plus MV. We used a two-hit murine model of ALI in which both inhaled LPS and MV were required for the development of hypoxemia, pulmonary neutrophil infiltration, and alveolar leakage. Nlrp3-deficent and Casp1-deficient mice had significantly diminished IL-1ß levels in bronchoalveolar lavage fluid, and were specifically protected from hypoxemia, despite similar alveolar neutrophil infiltration and leakage. The IL-1 receptor antagonist, Anakinra, significantly improved the specific development of hypoxemia without significant effects on neutrophil infiltration or alveolar leakage. MV resulted in increased bronchoalveolar lavage extracellular ATP and alveolar macrophage apoptosis as triggers of NLRP3 inflammasome activation. NLRP3 inflammasome activation and IL-1ß production play a key role in ALI caused by the combination of LPS and MV, particularly in the hypoxemia associated with acute respiratory distress syndrome. Blocking IL-1 signaling in this model specifically ameliorates hypoxemia, without affecting neutrophil infiltration and alveolar leakage, disassociating these readouts of ALI. MV causes alveolar macrophage apoptosis, a key step in the activation of NLRP3 inflammasome and production of IL-1ß.


Asunto(s)
Lesión Pulmonar Aguda/metabolismo , Proteínas Portadoras/metabolismo , Hipoxia/inmunología , Infiltración Neutrófila/inmunología , Lesión Pulmonar Aguda/inmunología , Animales , Caspasa 1/inmunología , Caspasa 1/metabolismo , Modelos Animales de Enfermedad , Inflamación/inmunología , Interleucina-1/inmunología , Interleucina-1/metabolismo , Interleucina-1beta/inmunología , Interleucina-1beta/metabolismo , Lipopolisacáridos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteína con Dominio Pirina 3 de la Familia NLR , Receptores de Interleucina-1/inmunología , Receptores de Interleucina-1/metabolismo , Respiración Artificial/efectos adversos , Respiración Artificial/métodos , Transducción de Señal/inmunología
11.
Biol Chem ; 395(10): 1173-8, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24633750

RESUMEN

Angiotensin-converting enzyme (ACE) plays an important role in blood pressure control. ACE also has effects on renal function, reproduction, hematopoiesis, and several aspects of the immune response. ACE 10/10 mice overexpress ACE in monocytic cells; macrophages from ACE 10/10 mice demonstrate increased polarization toward a proinflammatory phenotype. As a result, ACE 10/10 mice have a highly effective immune response following challenge with melanoma, bacterial infection, or Alzheimer disease. As shown in ACE 10/10 mice, enhanced monocytic function greatly contributes to the ability of the immune response to defend against a wide variety of antigenic and non-antigenic challenges.


Asunto(s)
Células Precursoras de Granulocitos/enzimología , Células Precursoras de Granulocitos/inmunología , Inmunidad Celular/genética , Peptidil-Dipeptidasa A/biosíntesis , Peptidil-Dipeptidasa A/genética , Animales , Ratones , Ratones Noqueados
12.
Curr Opin Nephrol Hypertens ; 23(2): 106-12, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24378774

RESUMEN

PURPOSE OF REVIEW: This review presents novel findings regarding the renal angiotensin-converting enzyme (ACE) and its role in blood pressure (BP) control. RECENT FINDINGS: The textbook flow diagram of the renin-angiotensin system (RAS) shows the pulmonary endothelium as the main source of the ACE that converts angiotensin I to angiotensin II. However, ACE is made in large quantities by the kidneys, which raises the important question of what precisely is the function of renal ACE? Recent studies in gene-targeted mice indicates that renal ACE plays a dominant role in regulating the response of the kidney to experimental hypertension. In particular, renal ACE and locally generated angiotensin II affect the activity of several key sodium transporters and the induction of sodium and water retention resulting in the elevation of BP. SUMMARY: New experimental data link the renal ACE/angiotensin II pathway and the local regulation of sodium transport as key elements in the development of hypertension.


Asunto(s)
Presión Sanguínea , Hipertensión/enzimología , Riñón/enzimología , Peptidil-Dipeptidasa A/metabolismo , Sistema Renina-Angiotensina , Angiotensina II/metabolismo , Animales , Agua Corporal/metabolismo , Humanos , Hipertensión/fisiopatología , Riñón/fisiopatología , Transducción de Señal , Sodio/metabolismo , Equilibrio Hidroelectrolítico , Desequilibrio Hidroelectrolítico/enzimología , Desequilibrio Hidroelectrolítico/fisiopatología
13.
Curr Hypertens Rep ; 16(9): 477, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25097114

RESUMEN

The existence of a complete and functional renin-angiotensin system along the nephron is widely recognized. However, its precise role in blood pressure control and, by extension, hypertension is still uncertain. While most investigators agree that overexpressing RAS components along the nephron results in hypertension, two important issues remain: whether the local RAS works as a separate entity or represents an extension of the systemic RAS and whether locally generated angiotensin II has specific renal effects on blood pressure that are distinct from systemic angiotensin II. This review addresses these issues while emphasizing the unique role of local angiotensin II in the response of the kidney to hypertensive stimuli and the induction of hypertension.


Asunto(s)
Angiotensina II/biosíntesis , Presión Sanguínea/fisiología , Hipertensión/metabolismo , Riñón/metabolismo , Sistema Renina-Angiotensina/fisiología , Animales , Humanos , Hipertensión/fisiopatología
14.
Curr Hypertens Rep ; 16(7): 444, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24792094

RESUMEN

While it is well known that angiotensin converting enzyme (ACE) plays an important role in blood pressure control, ACE also has effects on renal function, hematopoiesis, reproduction, and aspects of the immune response. ACE 10/10 mice overexpress ACE in myelomonocytic cells. Macrophages from these mice have an increased polarization towards a pro-inflammatory phenotype that results in a very effective immune response to challenge by tumors or bacterial infection. In a mouse model of Alzheimer's disease (AD), the ACE 10/10 phenotype provides significant protection against AD pathology, including reduced inflammation, reduced burden of the neurotoxic amyloid-ß protein and preserved cognitive function. Taken together, these studies show that increased myelomonocytic ACE expression in mice alters the immune response to better defend against many different types of pathologic insult, including the cognitive decline observed in an animal model of AD.


Asunto(s)
Enfermedad de Alzheimer/genética , Inhibidores de la Enzima Convertidora de Angiotensina/uso terapéutico , Hipertensión/metabolismo , Monocitos/enzimología , Peptidil-Dipeptidasa A/genética , Animales , Modelos Animales de Enfermedad , Humanos , Hipertensión/tratamiento farmacológico , Hipertensión/genética , Peptidil-Dipeptidasa A/metabolismo
15.
Nephron ; 2024 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-38301618

RESUMEN

INTRODUCTION: ob/ob mice are a leptin-deficient type 2 diabetes mellitus model, which, on a BTBR background, mimics glomerular pathophysiology of diabetic nephropathy (DN). Since leptin deficiency reduces blood pressure (BP), and endothelial nitric oxide synthase (eNOS) lowers BP and is kidney protective, we attempted to develop a more robust DN model by introducing eNOS deficiency in BTBR ob/ob mice. METHODS: Six experimental groups included littermate male and female BTBR ob/ob or wild-type for ob (control) as well as wild-type (WT), heterozygote (HET) or knockout (KO) for eNOS. Systolic BP (by automated tail-cuff) and GFR (by FITC sinistrin plasma kinetics) were determined in awake mice at 27-30 weeks of age followed by molecular and histological kidney analyses. RESULTS: Male and female ob/ob WT presented hyperglycemia and larger body and kidney weight, GFR, glomerular injury, and urine albumin to creatinine ratio (UACR) despite modestly lower BP vs control WT. These effects were associated with higher tubular injury score and renal mRNA expression of NGAL only in males, whereas female ob/ob WT unexpectedly had lower KIM-1 and COL1A1 expression vs control WT, indicating sex differences. HET for eNOS did not consistently alter BP or renal outcome in control or ob/ob. In comparison, eNOS KO increased BP (15-25 mmHg) and worsened renal markers of injury, inflammation and fibrosis, GFR, UACR, and survival rates, as observed in control and, more pronounced, in ob/ob mice and independent of sex. CONCLUSIONS: Deletion, but not heterozygosity, of eNOS raises blood pressure and aggravates nephropathy in BTBR ob/ob mice.

16.
FASEB J ; 26(5): 1821-30, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22302831

RESUMEN

Renal inflammation modulates angiotensinogen (AGT) production in renal proximal tubular cells (RPTCs) via inflammatory cytokines, including interleukin-6, tumor necrosis factor α, and interferon-γ (IFN-γ). Among these, the effects of IFN-γ on AGT regulation in RPTCs are incompletely delineated. This study aimed to elucidate mechanisms by which IFN-γ regulates AGT expression in RPTCs. RPTCs were incubated with or without IFN-γ up to 48 h. AGT expression, STAT1 and STAT3 activities, and SOCS1 expression were evaluated. RNA interference studies against STAT1, SOCS1, and STAT3 were performed to elucidate a signaling cascade. IFN-γ decreased AGT expression at 6 h (0.61±0.05, ratio to control) and 12 h (0.47±0.03). In contrast, longer exposure for 24 and 48 h increased AGT expression (1.76±0.18, EC(50)=3.4 ng/ml, and 1.45±0.08, respectively). IFN-γ treatment for 6 h strongly induced STAT1 phosphorylation and SOCS1 augmentation, and decreased STAT3 activity. However, STAT1 phosphorylation and SOCS1 augmentation waned at 24 h, while STAT3 activity increased. RNA interference studies revealed that activation of STAT1-SOCS1 axis decreased STAT3 activity. Thus, IFN-γ biphasically regulates AGT expression in RPTCs via STAT3 activity modulated by STAT1-SOCS1 axis, suggesting the STAT1-SOCS1 axis is important in IFN-γ-induced activation of the intrarenal renin-angiotensin system.


Asunto(s)
Angiotensinógeno/genética , Interferón gamma/fisiología , Quinasas Janus/metabolismo , Túbulos Renales Proximales/metabolismo , Factor de Transcripción STAT1/metabolismo , Factor de Transcripción STAT3/metabolismo , Proteínas Supresoras de la Señalización de Citocinas/fisiología , Animales , Secuencia de Bases , Línea Celular Transformada , Ensayo de Cambio de Movilidad Electroforética , Ensayo de Inmunoadsorción Enzimática , Túbulos Renales Proximales/citología , Fosforilación , Interferencia de ARN , ARN Interferente Pequeño , Ratas , Reacción en Cadena en Tiempo Real de la Polimerasa , Proteína 1 Supresora de la Señalización de Citocinas
17.
JCI Insight ; 8(3)2023 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-36752209

RESUMEN

Acute kidney failure and chronic kidney disease are global health issues steadily rising in incidence and prevalence. Animal models on a single genetic background have so far failed to recapitulate the clinical presentation of human nephropathies. Here, we used a simple model of folic acid-induced kidney injury in 7 highly diverse mouse strains. We measured plasma and urine parameters, as well as renal histopathology and mRNA expression data, at 1, 2, and 6 weeks after injury, covering the early recovery and long-term remission. We observed an extensive strain-specific response ranging from complete resistance of the CAST/EiJ to high sensitivity of the C57BL/6J, DBA/2J, and PWK/PhJ strains. In susceptible strains, the severe early kidney injury was accompanied by the induction of mitochondrial stress response (MSR) genes and the attenuation of NAD+ synthesis pathways. This is associated with delayed healing and a prolonged inflammatory and adaptive immune response 6 weeks after insult, heralding a transition to chronic kidney disease. Through a thorough comparison of the transcriptomic response in mouse and human disease, we show that critical metabolic gene alterations were shared across species, and we highlight the PWK/PhJ strain as an emergent model of transition from acute kidney injury to chronic disease.


Asunto(s)
Lesión Renal Aguda , Insuficiencia Renal Crónica , Humanos , Ratones , Animales , Ratones Endogámicos C57BL , NAD , Ratones Endogámicos DBA
18.
bioRxiv ; 2023 Apr 21.
Artículo en Inglés | MEDLINE | ID: mdl-37131719

RESUMEN

During fibroproliferation, protein-associated extracellular aldehydes are formed by the oxidation of lysine residues on extracellular matrix proteins to form the aldehyde allysine. Here we report three Mn(II)-based, small molecule magnetic resonance (MR) probes that contain α-effect nucleophiles to target allysine in vivo and report on tissue fibrogenesis. We used a rational design approach to develop turn-on probes with a 4-fold increase in relaxivity upon targeting. The effects of aldehyde condensation rate and hydrolysis kinetics on the performance of the probes to detect tissue fibrogenesis noninvasively in mouse models were evaluated by a systemic aldehyde tracking approach. We showed that for highly reversible ligations, off-rate was a stronger predictor of in vivo efficiency, enabling histologically validated, three-dimensional characterization of pulmonary fibrogenesis throughout the entire lung. The exclusive renal elimination of these probes allowed for rapid imaging of liver fibrosis. Reducing the hydrolysis rate by forming an oxime bond with allysine enabled delayed phase imaging of kidney fibrogenesis. The imaging efficacy of these probes, coupled with their rapid and complete elimination from the body, make them strong candidates for clinical translation.

19.
Am J Pathol ; 178(1): 434-41, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21224080

RESUMEN

Coronary artery disease in patients with hypertension is increasing worldwide and leads to severe cardiovascular complications. The cellular and molecular mechanisms that underlie this pathologic condition are not well understood. Experimental and clinical research indicates that immune cells and inflammation play a central role in the pathogenesis of cardiovascular diseases. Recently, it has been reported that CD4(+)CD25(+) regulatory T cells (Tregs) regulate heart fibrosis in hypertension. In this study, we determined the role of Tregs in coronary arteriolar endothelial dysfunction in angiotensin II-dependent hypertensive mice. Mice infused with angiotensin II had significantly increased blood pressure, as determined using telemetry, and apoptotic Treg numbers, as measured using flow cytometry. The mice displayed inflammation, assessed by macrophage activation/infiltration into coronary arterioles and the heart, and increased local tumor necrosis factor-α release, which participates in reduced coronary arteriolar endothelial-dependent relaxation in response to acetylcholine using an arteriograph. Hypertensive mice injected with Tregs isolated from control mice had significantly reduced macrophage activation and infiltration, reduced tumor necrosis factor-α release, and improved coronary arteriolar endothelium-dependent relaxation. Our novel data indicate that Tregs are important in the development of coronary arteriolar endothelial dysfunction in hypertension. These results suggest a new direction in the investigation of vascular disease in hypertension and could lead to a therapeutic strategy that involves immune system modulation using Tregs.


Asunto(s)
Vasos Coronarios/fisiopatología , Endotelio Vascular/fisiopatología , Hipertensión/fisiopatología , Linfocitos T Reguladores/inmunología , Animales , Arteriolas/inmunología , Arteriolas/fisiopatología , Vasos Coronarios/inmunología , Endotelio Vascular/inmunología , Hipertensión/inmunología , Recuento de Linfocitos , Macrófagos/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL
20.
J Am Soc Nephrol ; 22(3): 449-59, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21115616

RESUMEN

The contribution of the intrarenal renin-angiotensin system to the development of hypertension is incompletely understood. Here, we used targeted homologous recombination to generate mice that express angiotensin-converting enzyme (ACE) in the kidney tubules but not in other tissues. Mice homozygous for this genetic modification (ACE 9/9 mice) had low BP levels, impaired ability to concentrate urine, and variable medullary thinning. In accord with the ACE distribution, these mice also had reduced circulating angiotensin II and high plasma renin concentration but maintained normal kidney angiotensin II levels. In response to chronic angiotensin I infusions, ACE 9/9 mice displayed increased kidney angiotensin II, enhanced rate of urinary angiotensin II excretion, and development of hypertension. These findings suggest that intrarenal ACE-derived angiotensin II formation, even in the absence of systemic ACE, increases kidney angiotensin II levels and promotes the development of hypertension.


Asunto(s)
Angiotensina I/farmacología , Hipertensión/fisiopatología , Túbulos Renales/efectos de los fármacos , Túbulos Renales/metabolismo , Peptidil-Dipeptidasa A/metabolismo , Angiotensina I/administración & dosificación , Angiotensina II/metabolismo , Animales , Presión Sanguínea/efectos de los fármacos , Presión Sanguínea/fisiología , Peso Corporal/efectos de los fármacos , Peso Corporal/fisiología , Modelos Animales de Enfermedad , Femenino , Hipertensión/etiología , Hipertensión/patología , Infusiones Subcutáneas , Túbulos Renales/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Receptor de Angiotensina Tipo 1/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA