Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
Kidney Int ; 2024 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-38801992

RESUMEN

The mechanisms responsible for glomerular hemodynamic regulation with sodium-glucose co-transporter 2 (SGLT2) inhibitors in kidney disease due to type 2 diabetes remain unclear. Therefore, we investigated changes in glomerular hemodynamic function using an animal model of type 2 diabetes, treated with an SGLT2 inhibitor alone or in combination with a renin-angiotensin-aldosterone system inhibitor using male Zucker lean (ZL) and Zucker diabetic fatty (ZDF) rats. Afferent and efferent arteriolar diameter and single-nephron glomerular filtration rate (SNGFR) were evaluated in ZDF rats measured at 0, 30, 60, 90, or 120 minutes after the administration of a SGLT2 inhibitor (luseogliflozin). Additionally, we assessed these changes under the administration of the adenosine A1 receptor (A1aR) antagonist (8-cyclopentyl-1,3-dipropylxanthine), along with coadministration of luseogliflozin and an angiotensin II receptor blocker (ARB), telmisartan. ZDF rats had significantly increased SNGFR, and afferent and efferent arteriolar diameters compared to ZL rats, indicating glomerular hyperfiltration. Administration of luseogliflozin significantly reduced afferent vasodilatation and glomerular hyperfiltration, with no impact on efferent arteriolar diameter. Urinary adenosine levels were increased significantly in the SGLT2 inhibitor group compared to the vehicle group. A1aR antagonism blocked the effect of luseogliflozin on kidney function. Co-administration of the SGLT2 inhibitor and ARB decreased the abnormal expansion of glomerular afferent arterioles, whereas the efferent arteriolar diameter was not affected. Thus, regulation of afferent arteriolar vascular tone via the A1aR pathway is associated with glomerular hyperfiltration in type 2 diabetic kidney disease.

2.
FASEB J ; 37(9): e23129, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37606578

RESUMEN

During peritoneal dialysis (PD), the peritoneum is exposed to a bioincompatible dialysate, deteriorating the tissue and limiting the long-term effectiveness of PD. Peritoneal fibrosis is triggered by chronic inflammation induced by a variety of stimuli, including peritonitis. Exposure to PD fluid alters peritoneal macrophages phenotype. Inflammasome activation triggers chronic inflammation. First, it was determined whether inflammasome activation causes peritoneal deterioration. In the in vivo experiments, the increased expression of the inflammasome components, caspase-1 activity, and concomitant overproduction of IL-1ß and IL-18 were observed in a mouse model of peritoneal fibrosis. ASC-positive and F4/80-positive cells colocalized in the subperitoneal mesothelial cell layer. These macrophages expressed high CD44 levels indicating that the CD44-positive macrophages contribute to developing peritoneal deterioration. Furthermore, intravital imaging of the peritoneal microvasculature demonstrated that the circulating CD44-positive leukocytes may contribute to peritoneal fibrosis. Bone marrow transplantation in ASC-deficient mice suppressed inflammasome activation, thereby attenuating peritoneal fibrosis in a high glucose-based PD solution-injected mouse model. Our results suggest inflammasome activation in CD44-positive macrophages may be involved in developing peritoneal fibrosis. The inflammasome-derived pro-inflammatory cytokines might therefore serve as new biomarkers for developing encapsulating peritoneal sclerosis.


Asunto(s)
Fibrosis Peritoneal , Peritonitis , Animales , Ratones , Peritoneo , Inflamasomas , Modelos Animales de Enfermedad , Inflamación
3.
Kidney Int ; 101(3): 551-562, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34843756

RESUMEN

Heart failure is frequently accompanied by kidney failure and co-incidence of these organ failures worsens the mortality in patients with heart failure. Recent clinical observations revealed that increased kidney venous pressure, rather than decreased cardiac output, causes the deterioration of kidney function in patients with heart failure. However, the underlying pathophysiology is unknown. Here, we found that decreased blood flow velocity in peritubular capillaries by kidney congestion and upregulation of endothelial nuclear factor-κB (NF-κB) signaling synergistically exacerbate kidney injury. We generated a novel mouse model with unilateral kidney congestion by constriction of the inferior vena cava between kidney veins. Intravital imaging highlighted the notable dilatation of peritubular capillaries and decreased kidney blood flow velocity in the congestive kidney. Damage after ischemia reperfusion injury was exacerbated in the congestive kidney and accumulation of polymorphonuclear leukocytes within peritubular capillaries was noted at the acute phase after injury. Similar results were obtained in vitro, in which polymorphonuclear leukocytes adhesion on activated endothelial cells was decreased in flow velocity-dependent manner but cancelled by inhibition of NF-κB signaling. Pharmacological inhibition of NF-κB for the mice subjected by both kidney congestion and ischemia reperfusion injury ameliorated the accumulation of polymorphonuclear leukocytes and subsequent exacerbation of kidney injury. Thus, our study demonstrates the importance of decreased blood flow velocity accompanying activated NF-κB signaling in aggravation of kidney injury. Hence, inhibition of NF-κB signaling may be a therapeutic candidate for the vicious cycle between heart and kidney failure with increased kidney venous pressure.


Asunto(s)
Lesión Renal Aguda , Daño por Reperfusión , Lesión Renal Aguda/terapia , Animales , Células Endoteliales , Humanos , Riñón , Ratones , FN-kappa B , Daño por Reperfusión/complicaciones
4.
Int J Mol Sci ; 22(17)2021 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-34502177

RESUMEN

Chronic kidney disease is a common problem in the elderly and is associated with increased mortality. We have reported on the role of nitric oxide, which is generated from endothelial nitric oxide synthase (eNOS), in the progression of aged kidneys. To elucidate the role of endothelial dysfunction and the lack of an eNOS-NO pathway in ageing kidneys, we conducted experiments using eNOS and ASC-deficient mice. C57B/6 J mice (wild type (WT)), eNOS knockout (eNOS KO), and ASC knockout (ASC KO) mice were used in the present study. Then, eNOS/ASC double-knockout (eNOS/ASC DKO) mice were generated by crossing eNOS KO and ASC KO mice. These mice were sacrificed at 17-19 months old. The Masson positive area and the KIM-1 positive area tended to increase in eNOS KO mice, compared with WT mice, but not eNOS/ASC DKO mice. The COX-positive area was significantly reduced in eNOS KO mice, compared with WT and eNOS/ASC DKO mice. To determine whether inflammasomes were activated in infiltrating macrophages, the double staining of IL-18 and F4/80 was performed. IL-18 and F4/80 were found to be co-localised in the tubulointerstitial areas. Inflammasomes play a pivotal role in inflammaging in ageing kidneys. Furthermore, inflammasome activation may accelerate cellular senescence via mitochondrial dysfunction. The importance of endothelial function as a regulatory mechanism suggests that protection of endothelial function may be a potential therapeutic target.


Asunto(s)
Envejecimiento , Endotelio/fisiopatología , Inflamasomas , Riñón/fisiopatología , Mitocondrias/metabolismo , Óxido Nítrico Sintasa de Tipo III/metabolismo , Animales , Endotelio/enzimología , Endotelio/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Mitocondrias/fisiología , Óxido Nítrico/metabolismo , Enfermedades Vasculares/fisiopatología
5.
Circulation ; 140(4): 303-315, 2019 07 23.
Artículo en Inglés | MEDLINE | ID: mdl-30773020

RESUMEN

BACKGROUND: Sodium glucose cotransporter 2 inhibitors may reduce kidney hyperfiltration, thereby preventing diabetic kidney disease progression, which may in turn reduce cardiovascular risk, including heart failure. However, the mechanisms that regulate renal function responses to sodium glucose cotransporter 2 inhibition are not yet fully understood. We explored the renal protective effects of sodium glucose cotransporter 2 inhibition with empagliflozin, with a focus on glomerular hemodynamic effects and tubuloglomerular feedback using in vivo multiphoton microscopy imaging techniques. METHODS: C57BL/6 mice and spontaneously diabetic Ins2+/Akita mice were studied. The mice were treated with empagliflozin (20 mg·kg-1·d-1) and insulin for 4 weeks, and the single-nephron glomerular filtration rate was measured using multiphoton microscope. A neuronal nitric oxide synthase inhibitor (7-nitroindazole, 20 mg·kg-1·d-1) or a cyclooxygenase-2 inhibitor (SC58236, 6 mg/L), or an A1 adenosine receptor antagonist (8-cyclopentyl-1,3-dipropylxanthine, 1 mg·kg-1·d-1) was administered to elucidate the mechanisms of tubuloglomerular feedback signaling and single-nephron glomerular filtration rate regulation. RESULTS: The urinary excretion of adenosine, nitric oxide metabolites, and the prostanoid prostaglandin E2 was also quantified. The single-nephron glomerular filtration rate in the Ins2+/Akita group was higher than in controls (C57BL/6; 4.9±1.3 nL/min versus Ins2+/Akita; 15.8±6.8 nL/min) and lower in Ins2+/Akita /empagliflozin to 8.0±3.3 nL/min (P<0.01). In vivo imaging also revealed concomitant afferent arteriolar dilation (P<0.01) and increased glomerular permeability of albumin in the Ins2+/Akita group. Empagliflozin ameliorated these changes (P<0.01). Urinary adenosine excretion in the Ins2+/Akita/empagliflozin group was higher than in Ins2+/Akita (Ins2+/Akita; 3.4±1.4 nmol/d, Ins2+/Akita/empagliflozin; 11.2±3.0 nmol/d, P<0.05), whereas nitric oxide metabolites and prostaglandin E2 did not differ. A1 adenosine receptor antagonism, but not neuronal nitric oxide synthase or cyclooxygenase-2 inhibition, blocked the effect of empagliflozin on renal function. Empagliflozin increased urinary adenosine excretion and reduced hyperfiltration via afferent arteriolar constriction, effects that were abolished by A1 adenosine receptor blockade. CONCLUSIONS: Adenosine/A1 adenosine receptor pathways play a pivotal role in the regulation of the single-nephron glomerular filtration rate via tubuloglomerular feedback mechanisms in response to sodium glucose cotransporter 2 inhibition, which may contribute to renal and cardiovascular protective effects reported in clinical trials.


Asunto(s)
Diabetes Mellitus Experimental/diagnóstico por imagen , Diabetes Mellitus Experimental/tratamiento farmacológico , Técnicas In Vitro/métodos , Riñón/fisiopatología , Inhibidores del Cotransportador de Sodio-Glucosa 2/uso terapéutico , Animales , Compuestos de Bencidrilo/farmacología , Compuestos de Bencidrilo/uso terapéutico , Glucósidos/farmacología , Glucósidos/uso terapéutico , Humanos , Masculino , Ratones , Inhibidores del Cotransportador de Sodio-Glucosa 2/farmacología
6.
Curr Opin Nephrol Hypertens ; 29(1): 112-118, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31725011

RESUMEN

PURPOSE OF REVIEW: Emerging data have demonstrated that sodium-glucose cotransporter-2 (SGLT2) inhibitors prevent cardiovascular events, especially heart failure-associated endpoints. Cardiovascular outcome trials have also suggested their renoprotective effects. One large clinical trial investigated renal primary endpoints and demonstrated that SGLT2 inhibitors slowed the progression of diabetic kidney disease (DKD). This review summarizes clinical trial data on renal outcomes and discusses potential underlying mechanisms. RECENT FINDINGS: The EMPA-REG, CANVAS, and DECLARE-TIMI 58 studies revealed that SGLT2 inhibitors reduce the risk of cardiovascular events and concomitantly suggested that these drugs slow the progression of kidney disease in type 2 diabetes. The CREDENCE trial on patients with high-risk type 2 diabetes and chronic kidney disease demonstrated that canagliflozin treatment reduced the relative risk of a composite outcome, including end-stage kidney disease, serum creatinine doubling, and renal/cardiovascular death, by 30% in these patients. Animal experiments revealed that oxidative stress, inflammation, fibrosis, and tubuloglomerular feedback are underlying renoprotective mechanisms behind SGLT2 inhibitors. SUMMARY: Recent clinical trials have established the renoprotective effects of SGLT2 inhibitors. Further investigations on mechanisms of these renoprotective effects will provide deeper insights and understanding of pathogenetic properties of DKD.


Asunto(s)
Diabetes Mellitus Tipo 2/tratamiento farmacológico , Hipoglucemiantes/farmacología , Riñón/efectos de los fármacos , Inhibidores del Cotransportador de Sodio-Glucosa 2/farmacología , Animales , Nefropatías Diabéticas/tratamiento farmacológico , Nefropatías Diabéticas/etiología , Humanos , Inhibidores del Cotransportador de Sodio-Glucosa 2/uso terapéutico
7.
FASEB J ; 33(11): 12253-12263, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31431054

RESUMEN

Multiple clinical studies have shown that bardoxolone methyl, a potent activator of nuclear factor erythroid 2-related factor 2 (Nrf2), is effective in increasing glomerular filtration rate in patients with chronic kidney disease. However, whether an Nrf2 activator can protect tubules from proteinuria-induced tubular damage via anti-inflammatory and antioxidative stress mechanisms is unknown. Using an Institute of Cancer Research-derived glomerulonephritis (ICGN) mouse model of nephrosis, we examined the effects of dihydro-CDDO-trifluoroethyl amide (dh404), a rodent-tolerable bardoxolone methyl analog, in protecting the tubulointerstitium; dh404 markedly suppressed tubular epithelial cell damage in the renal interstitium of ICGN mice. The tubular epithelial cells of ICGN mice showed a decrease in the size and number of mitochondria, as well as the breakdown of the crista structure, whereas the number and ultrastructure of mitochondria were maintained by the dh404 treatment. To further determine the effect of dh404 on mitochondrial function, we used human proximal tubular cells in vitro. Stimulation with albumin and free fatty acid increased mitochondrial reactive oxygen species (ROS). However, dh404 administration diminished mitochondrial ROS. Our data show that dh404 significantly reduced proteinuria-induced tubular cell mitochondrial damage, suggesting that improved redox balance and mitochondrial function and suppression of inflammation underlie the cytoprotective mechanism of Nrf2 activators, including bardoxolone methyl, in diabetic kidney disease.-Nagasu, H., Sogawa, Y., Kidokoro, K., Itano, S., Yamamoto, T., Satoh, M., Sasaki, T., Suzuki, T., Yamamoto, M., Wigley, W. C., Proksch, J. W., Meyer, C. J., Kashihara, N. Bardoxolone methyl analog attenuates proteinuria-induced tubular damage by modulating mitochondrial function.


Asunto(s)
Túbulos Renales Proximales/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Ácido Oleanólico/análogos & derivados , Proteinuria/complicaciones , Animales , Células Cultivadas , Humanos , Túbulos Renales Proximales/patología , Masculino , Ratones , Ratones Endogámicos ICR , Mitocondrias/fisiología , Factor 2 Relacionado con NF-E2/fisiología , Ácido Oleanólico/farmacología , Especies Reactivas de Oxígeno/metabolismo
8.
Nephrol Dial Transplant ; 35(5): 773-781, 2020 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-32221606

RESUMEN

BACKGROUND: Long-term exposure to bioincompatible peritoneal dialysate causes the loss of mesothelial cells and accumulation of matrix proteins, leading to an increase in the thickness of the submesothelial layer, thereby limiting the long-term effectiveness of peritoneal dialysis (PD). However, the detailed molecular mechanisms underlying the process of peritoneal fibrosis have not been clearly elucidated. Wnt/ß-catenin signaling pathway activation has been suggested to play a pivotal role in the development of organ fibrosis. Moreover, Klotho protein can regulate Wnt/ß-catenin signaling. We examined the role of Klotho protein in reducing peritoneal fibrosis by inhibiting Wnt/ß-catenin signaling. METHODS: The ß-catenin-activated transgenic (BAT) driving expression of nuclear ß-galactosidase reporter transgenic (BAT-LacZ) mice, the alpha-Klotho gene under control of human elongation factor 1 alpha promoter [Klotho transgenic (KLTG) and C57BL/6 background] and C57BL/6 mice [wild-type (WT)] were used. The mice received daily intraperitoneal (i.p.) injections of 4.25% glucose with lactate (PD solution) or saline as a control for 4 weeks. Other mice received daily i.p. injections of the same volume of saline (normal control). RESULTS: After exposure to PD, Wnt signal activation was observed on the peritoneal mesothelial cells in WT-PD mice. The peritoneal fibrosis was also accelerated in WT-PD mice. The protein expression of ß-catenin and Wnt-inducible genes were also remarkably increased in WT-PD mice. On the other hand, KLTG-PD mice attenuated activation of Wnt/ß-catenin signaling after exposure to PD and ameliorated the progression of peritoneal fibrosis. CONCLUSIONS: Overexpression of Klotho protein protects the peritoneal membrane through attenuation of the Wnt/ß-catenin signaling pathway. The availability of recombinant Klotho protein would provide a novel potential therapeutic target in peritoneal fibrosis.


Asunto(s)
Glucuronidasa/fisiología , Fibrosis Peritoneal/terapia , Proteínas Wnt/antagonistas & inhibidores , beta Catenina/antagonistas & inhibidores , Animales , Humanos , Proteínas Klotho , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Diálisis Peritoneal/efectos adversos , Fibrosis Peritoneal/etiología , Fibrosis Peritoneal/patología , Proteínas Wnt/metabolismo , beta Catenina/metabolismo
9.
Nephrology (Carlton) ; 24(1): 28-38, 2019 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-29068550

RESUMEN

AIM: Acute kidney injury (AKI) is associated with chronic kidney disease, as well as high mortality, but effective treatments for AKI are still lacking. A recent study reported the prevention of renal injury, such as ischemia-reperfusion injury, by 5-aminolevulinic acid (ALA), which induces an antioxidant effect. The current study aimed to investigate the effect of ALA in a rhabdomyolysis-induced mouse model of AKI created by intramuscular injection of 50% glycerol. METHODS: Rhabdomyolysis-induced AKI was induced by an intramuscular injection of glycerol (5 mL/kg body weight) into mice. Administration of ALA (30 mg/kg, by gavage) was started from 48 h before or 24 h after glycerol injection. The mice were sacrificed at 72 h after glycerol injection. The roles of nuclear factor erythroid 2-related factor 2 (Nrf2) and heme oxygenase-1 (HO-1), which is one of the Nrf2-related antioxidants, were further investigated through in vivo and in vitro methods. RESULTS: 5-aminolevulinic acid markedly reduced renal dysfunction and tubular damage in mice with rhabdomyolysis-induced AKI. ALA administration decreased oxidative stress, macrophage infiltration, and inflammatory cytokines and apoptosis. The expression of Nrf2 was upregulated by ALA administration. However, administration of Zinc protoporphyrin-9 (ZnPPIX) to inhibit HO-1 activity did not abolish these improvements by ALA. The expression of Nrf2-associated antioxidant factors other than HO-1 was also increased. CONCLUSION: These findings indicate that ALA exerts its antioxidant activity via Nrf2-associated antioxidant factors to provide a renoprotective effect against rhabdomyolysis-induced AKI.


Asunto(s)
Lesión Renal Aguda/prevención & control , Ácido Aminolevulínico/farmacología , Antioxidantes/farmacología , Túbulos Renales/efectos de los fármacos , Factor 2 Relacionado con NF-E2/agonistas , Rabdomiólisis/prevención & control , Lesión Renal Aguda/inducido químicamente , Lesión Renal Aguda/metabolismo , Lesión Renal Aguda/patología , Animales , Apoptosis/efectos de los fármacos , Células Cultivadas , Citocinas/metabolismo , Citoprotección , Modelos Animales de Enfermedad , Glicerol , Hemo-Oxigenasa 1/metabolismo , Humanos , Mediadores de Inflamación/metabolismo , Túbulos Renales/metabolismo , Túbulos Renales/patología , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Macrófagos/patología , Masculino , Ratones Endogámicos C57BL , Factor 2 Relacionado con NF-E2/genética , Factor 2 Relacionado con NF-E2/metabolismo , Estrés Oxidativo/efectos de los fármacos , Rabdomiólisis/inducido químicamente , Rabdomiólisis/metabolismo , Rabdomiólisis/patología , Transducción de Señal/efectos de los fármacos
10.
Lab Invest ; 96(1): 25-36, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26552047

RESUMEN

Increased generation of reactive oxygen species (ROS) is a common denominative pathogenic mechanism underlying vascular and renal complications in diabetes mellitus. Endothelial NAD(P)H oxidase is a major source of vascular ROS, and it has an important role in endothelial dysfunction. We hypothesized that activation of endothelial NAD(P)H oxidase initiates and worsens the progression of diabetic nephropathy, particularly in the development of albuminuria. We used transgenic mice with endothelial-targeted overexpression of the catalytic subunit of NAD(P)H oxidase, Nox2 (NOX2TG). NOX2TG mice were crossed with Akita insulin-dependent diabetic (Akita) mice that develop progressive hyperglycemia. We compared the progression of diabetic nephropathy in Akita versus NOX2TG-Akita mice. NOX2TG-Akita mice and Akita mice developed significant albuminuria above the baseline at 6 and 10 weeks of age, respectively. Compared with Akita mice, NOX2TG-Akita mice exhibited higher levels of NAD(P)H oxidase activity in glomeruli, developed glomerular endothelial perturbations, and attenuated expression of glomerular glycocalyx. Moreover, in contrast to Akita mice, the NOX2TG-Akita mice had numerous endothelial microparticles (blebs), as detected by scanning electron microscopy, and increased glomerular permeability. Furthermore, NOX2TG-Akita mice exhibited distinct phenotypic changes in glomerular mesangial cells expressing α-smooth muscle actin, and in podocytes expressing increased levels of desmin, whereas the glomeruli generated increased levels of ROS. In conclusion, activation of endothelial NAD(P)H oxidase in the presence of hyperglycemia initiated and exacerbated diabetic nephropathy characterized by the development of albuminuria. Moreover, ROS generated in the endothelium compounded glomerular dysfunctions by altering the phenotypes of mesangial cells and compromising the integrity of the podocytes.


Asunto(s)
Diabetes Mellitus Experimental/metabolismo , Nefropatías Diabéticas/metabolismo , NADPH Oxidasas/metabolismo , Animales , Glucemia/análisis , Diabetes Mellitus Experimental/enzimología , Nefropatías Diabéticas/enzimología , Femenino , Tasa de Filtración Glomerular/fisiología , Glomérulos Renales/química , Glomérulos Renales/metabolismo , Glomérulos Renales/patología , Masculino , Ratones , Podocitos/química , Especies Reactivas de Oxígeno/metabolismo
11.
Curr Opin Nephrol Hypertens ; 25(3): 168-73, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-27008595

RESUMEN

PURPOSE OF REVIEW: The review aims to provide a brief summary and evaluation of the current state of research that uses multiphoton fluorescence microscopy for intravital kidney imaging. RECENT FINDINGS: Direct visualization of the glomerular filter, proximal and distal tubule segments, and the renal vasculature in the living, intact kidney in zebrafish, mouse, and rat models with high temporal and spatial resolution provided new insights into the function of the normal and diseased kidney. New technical developments in fluorescence excitation and detection, in combination with transgenic animal models for cell function and fate mapping, and serial imaging of the same glomerulus in the same animal over several days further advanced the field of nephrology research, and the understanding of disease mechanisms. SUMMARY: Intravital multiphoton imaging has solved many critical technical barriers in kidney research and allowed the dynamic portrayal of the structure and function of various renal cell types in vivo. It has become a widely used research technique, with significant past achievements, and tremendous potential for future development and applications for the study and better understanding of kidney diseases.


Asunto(s)
Microscopía Intravital , Enfermedades Renales/diagnóstico , Riñón/patología , Microscopía de Fluorescencia por Excitación Multifotónica , Animales , Células Epiteliales/metabolismo , Células Epiteliales/patología , Humanos , Riñón/irrigación sanguínea , Enfermedades Renales/patología , Microscopía de Fluorescencia por Excitación Multifotónica/métodos , Modelos Animales
12.
Kidney Int ; 88(1): 44-51, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25738253

RESUMEN

Intravital imaging using multiphoton microscopy (MPM) has become an increasingly popular and widely used experimental technique in kidney research over the past few years. MPM allows deep optical sectioning of the intact, living kidney tissue with submicron resolution, which is unparalleled among intravital imaging approaches. MPM has solved a long-standing critical technical barrier in renal research to study several complex and inaccessible cell types and anatomical structures in vivo in their native environment. Comprehensive and quantitative kidney structure and function MPM studies helped our better understanding of the cellular and molecular mechanisms of the healthy and diseased kidney. This review summarizes recent in vivo MPM studies with a focus on the glomerulus and the filtration barrier, although select, glomerulus-related renal vascular and tubular functions are also mentioned. The latest applications of serial MPM of the same glomerulus in vivo, in the intact kidney over several days, during the progression of glomerular disease are discussed. This visual approach, in combination with genetically encoded fluorescent markers of cell lineage, has helped track the fate and function (e.g., cell calcium changes) of single podocytes during the development of glomerular pathologies, and provided visual proof for the highly dynamic, rather than static, nature of the glomerular environment. Future intravital imaging applications have the promise to further push the limits of optical microscopy, and to advance our understanding of the mechanisms of kidney injury. Also, MPM will help to study new mechanisms of tissue repair and regeneration, a cutting-edge area of kidney research.


Asunto(s)
Barrera de Filtración Glomerular/anatomía & histología , Barrera de Filtración Glomerular/fisiología , Microscopía Intravital/métodos , Enfermedades Renales/patología , Microscopía de Fluorescencia por Excitación Multifotónica , Animales , Calcio/metabolismo , Movimiento Celular , Enfermedades Renales/fisiopatología , Ratones , Podocitos/fisiología , Ratas , Imagen de Lapso de Tiempo , Pez Cebra
13.
J Am Soc Nephrol ; 24(7): 1139-50, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23620395

RESUMEN

In diabetes, endothelial nitric oxide synthase (eNOS) produces superoxide anion rather than nitric oxide, referred to as "eNOS uncoupling," which may contribute to endothelial dysfunction, albuminuria, and diabetic nephropathy. Reduced levels of endothelium-derived tetrahydrobiopterin (BH4), an essential cofactor for eNOS, promote eNOS uncoupling. Accelerated degradation of guanosine triphosphate cyclohydrolase I (GTPCH I), the rate-limiting enzyme in BH4 biosynthesis, also occurs in diabetes, suggesting that GTPCH I may have a role in diabetic microvascular disease. Here, we crossed endothelium-dominant GTPCH I transgenic mice with Ins2(+/Akita) diabetic mice and found that endothelial overexpression of GTPCH I led to higher levels of intrarenal BH4 and lower levels of urinary albumin and reactive oxygen species compared with diabetic control mice. Furthermore, GTPCH I overexpression attenuated the hyperpermeability of macromolecules observed in diabetic control mice. In addition, we treated Ins2(+/Akita) mice with metformin, which activates AMP-activated protein kinase (AMPK) and thereby slows the degradation of GTPCH I; despite blood glucose levels that were similar to untreated mice, those treated with metformin had significantly less albuminuria. Similarly, in vitro, treating human glomerular endothelial cells with AMPK activators attenuated glucose-induced reductions in phospho-AMPK, GTPCH I, and coupled eNOS. Taken together, these data suggest that maintenance of endothelial GTPCH I expression and the resulting improvement in BH4 biosynthesis ameliorate diabetic nephropathy.


Asunto(s)
Biopterinas/análogos & derivados , Diabetes Mellitus Experimental/metabolismo , Nefropatías Diabéticas/metabolismo , GTP Ciclohidrolasa/metabolismo , Albuminuria/metabolismo , Animales , Biopterinas/metabolismo , Línea Celular , Endotelio Vascular/metabolismo , Humanos , Metformina/farmacología , Ratones , Óxido Nítrico Sintasa de Tipo III/metabolismo , Especies Reactivas de Oxígeno/metabolismo
14.
Nat Rev Nephrol ; 20(2): 101-119, 2024 02.
Artículo en Inglés | MEDLINE | ID: mdl-37857763

RESUMEN

Chronic kidney disease (CKD) is a major public health concern, underscoring a need to identify pathogenic mechanisms and potential therapeutic targets. Reactive oxygen species (ROS) are derivatives of oxygen molecules that are generated during aerobic metabolism and are involved in a variety of cellular functions that are governed by redox conditions. Low levels of ROS are required for diverse processes, including intracellular signal transduction, metabolism, immune and hypoxic responses, and transcriptional regulation. However, excess ROS can be pathological, and contribute to the development and progression of chronic diseases. Despite evidence linking elevated levels of ROS to CKD development and progression, the use of low-molecular-weight antioxidants to remove ROS has not been successful in preventing or slowing disease progression. More recent advances have enabled evaluation of the molecular interactions between specific ROS and their targets in redox signalling pathways. Such studies may pave the way for the development of sophisticated treatments that allow the selective control of specific ROS-mediated signalling pathways.


Asunto(s)
Estrés Oxidativo , Insuficiencia Renal Crónica , Humanos , Especies Reactivas de Oxígeno/metabolismo , Oxidación-Reducción , Transducción de Señal
15.
Lab Invest ; 93(3): 334-43, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23295649

RESUMEN

The development of interstitial fibrosis occurs with aging. Impaired angiogenesis, associated with progressive loss of the renal microvasculature, is thought to be a cause of age-related nephropathy. However, the mechanism of capillary loss in aging kidney has not been fully elucidated. Angiostatin is a kringle-containing fragment of plasminogen and is a potent inhibitor of angiogenesis in vivo. Whether angiostatin generation is increased in the aging kidney has not been investigated. We examined 4, 10, 16, and 24-month-old Sprague-Dawley rats for angiostatin production and found that angiostatin generation was increased in aged rats. The protein expression and the activity of cathepsin D-the enzyme for angiostatin production--were increased in aged rats. In the aging kidney, nitric oxide (NO) availability is decreased. To investigate the role of NO in angiostatin production, human umbilical vein endothelial cells were treated with L-NG-nitroarginine methyl ester (L-NAME). L-NAME-treated cells showed increased cathepsin D activity and angiostatin production. For in vivo experiments, 16- to 18-month-old rats were treated with L-NAME or molsidomine for 3 months. Angiostatin production was increased in L-NAME-treated kidney, accompanied by increased cathepsin D activity. In contrast, angiostatin production was decreased in molsidomine-treated kidney, accompanied by decreased cathepsin D activity. In conclusion, angiostatin generation by cathepsin D was increased in the aging rat kidney. Decreased NO production activated cathepsin D activity. Increased angiostatin production may be related to capillary loss and interstitial damage in the aging rat kidney.


Asunto(s)
Envejecimiento/metabolismo , Angiostatinas/biosíntesis , Riñón/metabolismo , Óxido Nítrico/metabolismo , Animales , Western Blotting , Catepsina D/metabolismo , Células Endoteliales de la Vena Umbilical Humana , Humanos , Inmunohistoquímica , Molsidomina , NG-Nitroarginina Metil Éster/farmacología , Ratas , Ratas Sprague-Dawley , Reacción en Cadena en Tiempo Real de la Polimerasa , Estadísticas no Paramétricas
16.
Kidney Int ; 83(4): 662-73, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23344476

RESUMEN

Proteinuria is an independent risk factor for progressive renal diseases because it initiates or aggravates tubulointerstitial injury. Clinically, females are less susceptible to progression of chronic kidney disease; however, the mechanisms underlying the renoprotective effect of estrogen receptor stimulation have yet to be clarified. Recently, inflammasome-dependent inflammatory responses were shown to be triggered by free fatty acids, and mitochondria-derived reactive oxygen species were shown to be required for this response. Albumin-bound free fatty acids trigger inflammasome activation through mitochondrial reactive oxygen species production in human proximal tubule epithelial cells in vitro, an effect inhibited by raloxifene. Female ICR-derived glomerulonephritic mice (mice with hereditary nephritic syndrome) were ovariectomized and treated with raloxifene, a selective estrogen receptor modulator. Ovariectomized mice showed activation of tubular inflammasomes and elevated levels of inflammasome-dependent cytokines. Raloxifene attenuated these changes ameliorating tubulointerstitial damage, reduced production of reactive oxygen species, averted morphological changes, and improved respiratory function in mitochondria. The expression of genes that encode rate-limiting enzymes in the mitochondrial ß-oxidation pathway was reduced by ovariectomy but enhanced by raloxifene. Thus, inflammasomes may be a novel and promising therapeutic target for proteinuria-induced renal injury.


Asunto(s)
Apoptosis/efectos de los fármacos , Glomerulonefritis/tratamiento farmacológico , Túbulos Renales Proximales/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Nefritis Hereditaria/tratamiento farmacológico , Estrés Oxidativo/efectos de los fármacos , Proteinuria/tratamiento farmacológico , Clorhidrato de Raloxifeno/farmacología , Moduladores Selectivos de los Receptores de Estrógeno/farmacología , Animales , Atrofia , Células Cultivadas , Citocinas/metabolismo , Modelos Animales de Enfermedad , Femenino , Fibrosis , Regulación de la Expresión Génica , Glomerulonefritis/genética , Glomerulonefritis/inmunología , Glomerulonefritis/metabolismo , Glomerulonefritis/patología , Humanos , Inflamasomas/efectos de los fármacos , Inflamasomas/inmunología , Mediadores de Inflamación/metabolismo , Túbulos Renales Proximales/inmunología , Túbulos Renales Proximales/metabolismo , Túbulos Renales Proximales/patología , Peroxidación de Lípido/efectos de los fármacos , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Ratones , Ratones Endogámicos ICR , Mitocondrias/inmunología , Mitocondrias/metabolismo , Mitocondrias/patología , FN-kappa B/metabolismo , Nefritis Hereditaria/genética , Nefritis Hereditaria/inmunología , Nefritis Hereditaria/metabolismo , Nefritis Hereditaria/patología , Ovariectomía , Oxidación-Reducción , Proteinuria/genética , Proteinuria/inmunología , Proteinuria/metabolismo , Proteinuria/patología
17.
Am J Physiol Renal Physiol ; 302(11): F1402-8, 2012 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-22378818

RESUMEN

Loss of functional nephrons associated with chronic kidney disease induces glomerular hyperfiltration and compensatory renal hypertrophy. We hypothesized that the endothelial nitric oxide synthase (eNOS) [soluble guanylate cyclase (sGC)] protein kinase G (PKG) pathway plays an important role in compensatory renal hypertrophy after unilateral nephrectomy. Analysis of mice subjected to unilateral nephrectomy showed increases in kidney weight-to-body weight and total protein-to-DNA ratios in wild-type but not eNOS knockout (eNOSKO) mice. Serum creatinine and blood urea nitrogen increased after nephrectomy in eNOSKO but not in wild-type mice. Furthermore, Bay 41-2272, an sGC stimulator, induced compensatory renal hypertrophy in eNOSKO mice and rescued renal function. The NO donor S-nitrosoglutathione (GSNO) and Bay 41-2272 stimulated PKG activity and induced phosphorylation of Akt protein in human proximal tubular cells. GSNO also induced phosphorylation of eukaryotic initiation factor 4E-binding protein and ribosomal protein S6. Our results highlight the importance of the eNOS-NO-PKG pathway in compensatory renal hypertrophy and suggest that reduced eNOS-NO bioavailability due to endothelial dysfunction is the underlying mechanism of failure of compensatory hypertrophy and acceleration of progressive renal dysfunction.


Asunto(s)
Lesión Renal Aguda/patología , Endotelio/fisiología , Riñón/patología , Óxido Nítrico Sintasa de Tipo III/metabolismo , Animales , Western Blotting , Recuento de Células , Proteínas Quinasas Dependientes de GMP Cíclico/metabolismo , ADN/metabolismo , Endotelio/enzimología , Hipertrofia , Riñón/enzimología , Túbulos Renales Proximales/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Nefrectomía , Óxido Nítrico/fisiología , Donantes de Óxido Nítrico/farmacología , Óxido Nítrico Sintasa de Tipo III/genética , Adhesión en Parafina , Biosíntesis de Proteínas , ARN/biosíntesis , ARN/aislamiento & purificación , Circulación Renal , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Serina-Treonina Quinasas TOR/metabolismo
18.
Kidney Blood Press Res ; 35(6): 549-57, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22890154

RESUMEN

BACKGROUND/AIMS: The immunosuppressive drug tacrolimus (FK506) is used clinically to reduce the rejection rate in patients with kidney transplantation; however, the resultant nephrotoxicity remains a serious problem. In the present study we attempted to elucidate the mechanisms of glomerular injury induced by FK506 and the renoprotective effects of the angiotensin II receptor blocker telmisartan. METHODS: Seven-week-old male Wistar rats were divided into three groups: vehicle group, FK506 group, and FK506 + telmisartan group. After 8 weeks, we assessed kidney function and renal morphological changes including oxidative stress. We also assessed the effect of FK506 in human glomerular endothelial cells (hGECs) with regard to reactive oxygen species (ROS). RESULTS: FK506 induced ROS production via activation of NAD(P)H oxidase in the glomeruli. Expression of ICAM mRNA was increased in glomeruli from the FK506 group. These effects resulted in macrophage infiltration into the glomeruli. FK506 directly promoted NAD(P)H oxidase activity and accelerated production of ROS in hGECs. Conversely, cotreatment with telmisartan inhibited both NAD(P)H oxidase activity and production of ROS. CONCLUSION: These findings suggest that glomerular injury resulting from FK506 is caused by oxidative stress mediated by activation of NAD(P)H oxidase and that telmisartan exerts a renoprotective effect via antioxidative activity.


Asunto(s)
Endotelio Vascular/metabolismo , Inmunosupresores/toxicidad , Glomérulos Renales/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Tacrolimus/toxicidad , Animales , Células Cultivadas , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/patología , Humanos , Inflamación/inducido químicamente , Inflamación/metabolismo , Inflamación/patología , Glomérulos Renales/efectos de los fármacos , Glomérulos Renales/patología , Masculino , Ratas , Ratas Wistar
19.
Transplantation ; 106(2): 280-288, 2022 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-33908383

RESUMEN

BACKGROUND: Neutrophils play a major role in ischemia/reperfusion injury (IRI) in renal transplantation and acute kidney injury. However, it has been difficult to observe changes in neutrophil dynamics over time in living mice kidney. We investigate neutrophil dynamics in IRI in living mice using novel in vivo multiphoton microscope imaging techniques and characterize the renoprotective effects of a selective phosphodiesterase 5 inhibitor, tadalafil. METHODS: Wild-type and endothelial nitric oxide synthase knockout mice, a model of endothelial dysfunction, were used to establish in vivo real-time imaging in living mouse kidneys. Neutrophils were labeled green with Ly-6G monoclonal antibody, and plasma flow was labeled red with BSA. Tadalafil was administered orally 1 h before surgery. Both kidney pedicles were reperfused after 37°C warm ischemia for 45 min. RESULTS: Our novel approach revealed that neutrophils were trapped in glomerulus within a few minutes after reperfusion. They gradually increased over time and infiltrated neutrophils were observed in the tubular lumen and peritubular capillary. The neutrophils were clearly visualized rolling on peritubular capillary plexus at 3 µm/min. The administration of tadalafil significantly reduced neutrophil influx into the glomerulus in both wild-type and endothelial nitric oxide synthase knockout mice. Reduced neutrophil infiltration in tadalafil groups, which was confirmed by flow cytometry, resulted in histopathologically decreased tubular injury. The expression of vascular cell adhesion molecule 1 and kidney injury molecule 1 was partially prevented by tadalafil. CONCLUSIONS: Use of a novel technique contributed to elucidation of neutrophil dynamics after reperfusion. Tadalafil has a potential for inhibiting neutrophil infiltration in renal IRI.


Asunto(s)
Lesión Renal Aguda , Daño por Reperfusión , Lesión Renal Aguda/tratamiento farmacológico , Lesión Renal Aguda/prevención & control , Animales , Isquemia , Riñón , Ratones , Neutrófilos/metabolismo , Reperfusión , Daño por Reperfusión/metabolismo , Tadalafilo/metabolismo , Tadalafilo/farmacología
20.
Biochem Biophys Res Commun ; 407(1): 39-43, 2011 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-21354104

RESUMEN

The klotho gene is highly expressed in the distal convoluted tubule of the kidney, while its encoded protein has many physiological and pathophysiological renal roles. We investigated the effect of klotho protein on physiological compensatory renal hypertrophy after nephrectomy in klotho transgenic (KLTG) mice. Renal hypertrophy was suppressed in KLTG mice compared with wild-type mice, and this was associated with suppression of insulin growth factor-1 (IGF-1) signaling by klotho protein. In vitro, IGF-1 signaling was suppressed in human proximal tubular cells transfected with the klotho plasmid. Our data suggest that klotho modulates compensatory renal hypertrophy after nephrectomy via suppression of the IGF-1 signaling pathway, indicating a novel physiological role for klotho protein in the kidney.


Asunto(s)
Glucuronidasa/fisiología , Factor I del Crecimiento Similar a la Insulina/antagonistas & inhibidores , Riñón/patología , Animales , Línea Celular , Glucuronidasa/genética , Humanos , Hipertrofia/genética , Hipertrofia/patología , Factor I del Crecimiento Similar a la Insulina/metabolismo , Riñón/fisiopatología , Glomérulos Renales/patología , Glomérulos Renales/fisiopatología , Túbulos Renales Proximales/metabolismo , Túbulos Renales Proximales/patología , Proteínas Klotho , Ratones , Ratones Transgénicos , NADPH Oxidasas/metabolismo , Nefrectomía , Transducción de Señal , Serina-Treonina Quinasas TOR/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA