Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
J Am Soc Nephrol ; 33(12): 2153-2173, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36198430

RESUMEN

BACKGROUND: The signaling molecule stimulator of IFN genes (STING) was identified as a crucial regulator of the DNA-sensing cyclic GMP-AMP synthase (cGAS)-STING pathway, and this signaling pathway regulates inflammation and energy homeostasis under conditions of obesity, kidney fibrosis, and AKI. However, the role of STING in causing CKD, including diabetic kidney disease (DKD) and Alport syndrome, is unknown. METHODS: To investigate whether STING activation contributes to the development and progression of glomerular diseases such as DKD and Alport syndrome, immortalized human and murine podocytes were differentiated for 14 days and treated with a STING-specific agonist. We used diabetic db/db mice, mice with experimental Alport syndrome, C57BL/6 mice, and STING knockout mice to assess the role of the STING signaling pathway in kidney failure. RESULTS: In vitro, murine and human podocytes express all of the components of the cGAS-STING pathway. In vivo, activation of STING renders C57BL/6 mice susceptible to albuminuria and podocyte loss. STING is activated at baseline in mice with experimental DKD and Alport syndrome. STING activation occurs in the glomerular but not the tubulointerstitial compartment in association with autophagic podocyte death in Alport syndrome mice and with apoptotic podocyte death in DKD mouse models. Genetic or pharmacologic inhibition of STING protects from progression of kidney disease in mice with DKD and Alport syndrome and increases lifespan in Alport syndrome mice. CONCLUSION: The activation of the STING pathway acts as a mediator of disease progression in DKD and Alport syndrome. Targeting STING may offer a therapeutic option to treat glomerular diseases of metabolic and nonmetabolic origin or prevent their development, progression, or both.


Asunto(s)
Nefropatías Diabéticas , Nefritis Hereditaria , Podocitos , Ratones , Humanos , Animales , Nefritis Hereditaria/genética , Nefritis Hereditaria/metabolismo , Ratones Endogámicos C57BL , Podocitos/metabolismo , Proteinuria/metabolismo , Nefropatías Diabéticas/genética , Nefropatías Diabéticas/metabolismo , Ratones Noqueados , Nucleotidiltransferasas/metabolismo
2.
Am J Physiol Cell Physiol ; 322(3): C468-C481, 2022 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-35108119

RESUMEN

Advances in the understanding of lipid droplet biology have revealed essential roles for these organelles in mediating proper cellular homeostasis and stress response. Lipid droplets were initially thought to play a passive role in energy storage. However, recent studies demonstrate that they have substantially broader functions, including protection from reactive oxygen species, endoplasmic reticulum stress, and lipotoxicity. Dysregulation of lipid droplet homeostasis is associated with various pathologies spanning neurological, metabolic, cardiovascular, oncological, and renal diseases. This review provides an overview of the current understanding of lipid droplet biology in both health and disease.


Asunto(s)
Gotas Lipídicas , Metabolismo de los Lípidos , Estrés del Retículo Endoplásmico , Homeostasis , Gotas Lipídicas/metabolismo , Metabolismo de los Lípidos/fisiología
3.
Kidney Int ; 101(3): 454-456, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35190031

RESUMEN

Lipid dysmetabolism is emerging as an important contributor to diabetic kidney disease, suggesting that intrarenal lipid accumulation is detrimental to kidney function. This commentary discusses the finding by Yoshioka et al., connecting tubular lipotoxicity induced by an increase in locally produced lysophosphatidylcholine in patients with a fast progression of diabetic kidney disease, known as "fast decliner." Insight into the lipid species in the kidney may prove beneficial for the diagnosis and stratification of patients with diabetic kidney disease.


Asunto(s)
Diabetes Mellitus , Nefropatías Diabéticas , Nefropatías Diabéticas/etiología , Humanos , Riñón , Nefronas
4.
Biochem Biophys Res Commun ; 515(1): 72-76, 2019 07 12.
Artículo en Inglés | MEDLINE | ID: mdl-31128920

RESUMEN

Ischemic injury is a primary contributor to the initiation of renal tubular epithelial cell damage in sickle cell disease (SCD). In this study, we investigated the effects of bilateral ischemia-reperfusion injury, which is a common type of acute kidney injury (AKI), in male and female genetic mouse model of SCD. Bilateral occlusion of both renal hila for 21 min led to a significantly higher detection of established serum markers of AKI (creatinine, KIM-1 and NGAL) compared to sham-operated male SCD mice. Severe damage to the outer medullary tubules was determined in the ischemia-reperfision injury (IRI)-treated SCD male mice. In female SCD mice with a longer ischemic time (23 min), the serum markers of AKI were not as highly elevated compared to their male counterparts, and the extent of outer medullary tubular injury was less severe. To assess the potential benefit in the use of hydroxyurea (50 mg/kg IP) following bilateral renal IRI, we observed that the serum markers of AKI and the outer medullary tubular damage were markedly improved compared to male SCD mice that were not treated with hydroxyurea. In this study, we confirmed that male SCD mice were more susceptible to increased tubular damage and a loss in renal function compared to female SCD mice, and that hydroxyurea may partially prevent the extent of tubular injury following severe ischemia-reperfusion injury in SCD.


Asunto(s)
Lesión Renal Aguda/fisiopatología , Anemia de Células Falciformes/tratamiento farmacológico , Hidroxiurea/farmacología , Túbulos Renales/efectos de los fármacos , Daño por Reperfusión/fisiopatología , Lesión Renal Aguda/sangre , Anemia de Células Falciformes/sangre , Animales , Antidrepanocíticos/farmacología , Biomarcadores/sangre , Creatinina/sangre , Modelos Animales de Enfermedad , Femenino , Receptor Celular 1 del Virus de la Hepatitis A/sangre , Túbulos Renales/metabolismo , Túbulos Renales/patología , Lipocalina 2/sangre , Masculino , Ratones , Daño por Reperfusión/sangre
5.
J Pharmacol Exp Ther ; 364(2): 287-299, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29187590

RESUMEN

Ischemia-reperfusion injury (IRI) is a common cause of acute kidney injury (AKI), which is an increasing problem in the clinic and has been associated with elevated rates of mortality. Therapies to treat AKI are currently not available, so identification of new targets that can be modulated to ameliorate renal damage upon diagnosis of AKI is essential. In this study, a novel cannabinoid receptor 2 (CB2) agonist, SMM-295 [3'-methyl-4-(2-(thiophen-2-yl)propan-2-yl)biphenyl-2,6-diol], was designed, synthesized, and tested in vitro and in silico. Molecular docking of SMM-295 into a CB2 active-state homology model showed that SMM-295 interacts well with key amino acids to stabilize the active state. In human embryonic kidney 293 cells, SMM-295 was capable of reducing cAMP production with 66-fold selectivity for CB2 versus cannabinoid receptor 1 and dose-dependently increased mitogen-activated protein kinase and Akt phosphorylation. In vivo testing of the CB2 agonist was performed using a mouse model of bilateral IRI, which is a common model to mimic human AKI, where SMM-295 was immediately administered upon reperfusion of the kidneys after the ischemia episode. Histologic damage assessment 48 hours after reperfusion demonstrated reduced tubular damage in the presence of SMM-295. This was consistent with reduced plasma markers of renal dysfunction (i.e., creatinine and neutrophil gelatinase-associated lipocalin) in SMM-295-treated mice. Mechanistically, kidneys treated with SMM-295 were shown to have elevated activation of Akt with reduced terminal deoxynucleotidyl transferase-mediated digoxigenin-deoxyuridine nick-end labeling (TUNEL)-positive cells compared with vehicle-treated kidneys after IRI. These data suggest that selective CB2 receptor activation could be a potential therapeutic target in the treatment of AKI.


Asunto(s)
Compuestos de Bifenilo/farmacología , Células Epiteliales/efectos de los fármacos , Células Epiteliales/patología , Túbulos Renales/efectos de los fármacos , Túbulos Renales/patología , Receptor Cannabinoide CB2/agonistas , Daño por Reperfusión/patología , Tiofenos/farmacología , Animales , Compuestos de Bifenilo/química , Compuestos de Bifenilo/metabolismo , Compuestos de Bifenilo/uso terapéutico , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Simulación del Acoplamiento Molecular , Permeabilidad , Conformación Proteica , Receptor Cannabinoide CB2/química , Receptor Cannabinoide CB2/metabolismo , Daño por Reperfusión/tratamiento farmacológico , Solubilidad , Tiofenos/química , Tiofenos/metabolismo , Tiofenos/uso terapéutico
6.
Bioorg Med Chem ; 26(17): 4787-4796, 2018 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-30145051

RESUMEN

Pleuromutilin is a promising pharmacophore to design new antibacterial agents for Gram-positive bacteria. However, there are limited studies on the development of pleuromutilin analogues that inhibit growth of Mycobacterium tuberculosis (Mtb). In screening of our library of pleuromutilin derivatives, UT-800 (1) was identified to kill replicating- and non-replicating Mtb with the MIC values of 0.83 and 1.20 µg/mL, respectively. UT-800 also kills intracellular Mtb faster than rifampicin at 2× MIC concentrations. Pharmacokinetic studies indicate that 1 has an oral bioavailability with an average F-value of 27.6%. Pleuromutilin may have the potential to be developed into an orally administered anti-TB drug.


Asunto(s)
Antituberculosos/farmacología , Mycobacterium tuberculosis/efectos de los fármacos , Administración Oral , Animales , Antituberculosos/administración & dosificación , Antituberculosos/farmacocinética , Área Bajo la Curva , Disponibilidad Biológica , Células CACO-2 , Diterpenos/administración & dosificación , Diterpenos/química , Diterpenos/farmacocinética , Diterpenos/farmacología , Femenino , Semivida , Humanos , Concentración 50 Inhibidora , Ratones , Ratones Endogámicos BALB C , Pruebas de Sensibilidad Microbiana , Mycobacterium tuberculosis/crecimiento & desarrollo , Compuestos Policíclicos , Pleuromutilinas
7.
Am J Physiol Renal Physiol ; 312(4): F551-F555, 2017 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-27927651

RESUMEN

Ischemia-reperfusion injury (IRI) is a common cause of acute kidney injury leading to an induction of oxidative stress, cellular dysfunction, and loss of renal function. DNA damage, including oxidative base modifications and physical DNA strand breaks, is a consequence of renal IRI. Like many other organs in the body, a redundant and highly conserved set of endogenous repair pathways have evolved to selectively recognize the various types of cellular DNA damage and combat its negative effects on cell viability. Severe damage to the DNA, however, can trigger cell death and elimination of the injured tubular epithelial cells. In this minireview, we summarize the state of the current field of DNA damage and repair in the kidney and provide some expected and, in some cases, unexpected effects of IRI on DNA damage and repair in the kidney. These findings may be applicable to other forms of acute kidney injury and could provide new opportunities for renal research.


Asunto(s)
Lesión Renal Aguda/genética , Daño del ADN , Reparación del ADN , Túbulos Renales/metabolismo , Daño por Reperfusión/genética , Lesión Renal Aguda/metabolismo , Lesión Renal Aguda/patología , Animales , Muerte Celular , Proliferación Celular , Células Epiteliales/metabolismo , Células Epiteliales/patología , Humanos , Túbulos Renales/patología , Estrés Oxidativo , Daño por Reperfusión/metabolismo , Daño por Reperfusión/patología , Transducción de Señal
8.
Mol Pharmacol ; 86(4): 369-77, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25028481

RESUMEN

Heterotrimeric G-proteins play a crucial role in the control of renal epithelial cell function during homeostasis and in response to injury. In this report, G-protein ßγ subunit (Gßγ) dimer activity was evaluated during the process of tubular repair after renal ischemia-reperfusion injury (IRI) in male Sprague Dawley rats. Rats were treated with a small molecule inhibitor of Gßγ activity, gallein (30 or 100 mg/kg), 1 hour after reperfusion and every 24 hours for 3 additional days. After IRI, renal dysfunction was prolonged after the high-dose gallein treatment in comparison with vehicle treatment during the 7-day recovery period. Renal tubular repair in the outer medulla 7 days after IRI was significantly (P < 0.001) attenuated after treatment with high-dose gallein (100 mg/kg) in comparison with low-dose gallein (30 mg/kg), or the vehicle and fluorescein control groups. Gallein treatment significantly reduced (P < 0.05) the number of proliferating cell nuclear antigen-positive tubular epithelial cells at 24 hours after the ischemia-reperfusion phase in vivo. In vitro application of gallein on normal rat kidney (NRK-52E) proximal tubule cells significantly reduced (P < 0.05) S-phase cell cycle entry compared with vehicle-treated cells as determined by 5'-bromo-2'-deoxyuridine incorporation. Taken together, these data suggest that Gßγ signaling contributes to the maintenance and repair of renal tubular epithelium and may be a novel therapeutic target for the development of drugs to treat acute kidney injury.


Asunto(s)
Síndrome Cardiorrenal/tratamiento farmacológico , Subunidades beta de la Proteína de Unión al GTP/metabolismo , Subunidades gamma de la Proteína de Unión al GTP/metabolismo , Riñón/efectos de los fármacos , Daño por Reperfusión/metabolismo , Animales , Síndrome Cardiorrenal/metabolismo , Línea Celular , Movimiento Celular , Proliferación Celular , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Células Epiteliales/fisiología , Subunidades beta de la Proteína de Unión al GTP/antagonistas & inhibidores , Subunidades gamma de la Proteína de Unión al GTP/antagonistas & inhibidores , Riñón/metabolismo , Riñón/patología , Masculino , Multimerización de Proteína , Ratas , Ratas Sprague-Dawley , Daño por Reperfusión/tratamiento farmacológico , Xantenos/farmacología , Xantenos/uso terapéutico
9.
Nat Commun ; 12(1): 4662, 2021 08 02.
Artículo en Inglés | MEDLINE | ID: mdl-34341345

RESUMEN

Impaired cellular cholesterol efflux is a key factor in the progression of renal, cardiovascular, and autoimmune diseases. Here we describe a class of 5-arylnicotinamide compounds, identified through phenotypic drug discovery, that upregulate ABCA1-dependent cholesterol efflux by targeting Oxysterol Binding Protein Like 7 (OSBPL7). OSBPL7 was identified as the molecular target of these compounds through a chemical biology approach, employing a photoactivatable 5-arylnicotinamide derivative in a cellular cross-linking/immunoprecipitation assay. Further evaluation of two compounds (Cpd A and Cpd G) showed that they induced ABCA1 and cholesterol efflux from podocytes in vitro and normalized proteinuria and prevented renal function decline in mouse models of proteinuric kidney disease: Adriamycin-induced nephropathy and Alport Syndrome. In conclusion, we show that small molecule drugs targeting OSBPL7 reveal an alternative mechanism to upregulate ABCA1, and may represent a promising new therapeutic strategy for the treatment of renal diseases and other disorders of cellular cholesterol homeostasis.


Asunto(s)
Transportador 1 de Casete de Unión a ATP/metabolismo , Colesterol/metabolismo , Nefropatías Diabéticas/metabolismo , Compuestos Orgánicos/farmacología , Podocitos/metabolismo , Proteinuria/metabolismo , Receptores de Esteroides/antagonistas & inhibidores , Transportador 1 de Casete de Unión a ATP/genética , Animales , Transporte Biológico/efectos de los fármacos , Células Cultivadas , Modelos Animales de Enfermedad , Células HEK293 , Humanos , Riñón/efectos de los fármacos , Riñón/metabolismo , Riñón/patología , Ratones de la Cepa 129 , Ratones Noqueados , Estructura Molecular , Niacinamida/química , Niacinamida/farmacología , Compuestos Orgánicos/síntesis química , Compuestos Orgánicos/química , Podocitos/citología , Interferencia de ARN , Receptores de Esteroides/genética , Receptores de Esteroides/metabolismo , Células THP-1
11.
Sci Rep ; 7: 43196, 2017 03 03.
Artículo en Inglés | MEDLINE | ID: mdl-28256593

RESUMEN

Damage to renal tubular epithelial cells by genetic, environmental, or biological insults can initiate complex signaling mechanisms that promote kidney repair and functional recovery. In this study, we demonstrated that thyroid receptor interacting protein 13 (TRIP13) is a critical modulator of tubular epithelial cell repair following ischemia-reperfusion injury (IRI), a common type of renal stressor. In Trip13Gt/Gthypomorph mice treated with unilateral renal IRI, persistent tubular epithelial cell damage was determined in the IRI-treated kidney throughout the 168 hours of experimental period compared to the contralateral kidneys. The damaged epithelial cells were associated with increased levels of DNA damage (É£H2AX) and apoptotic markers (p53, cleaved caspase-7, and TUNEL-positive cells). Correspondingly, TRIP13 was found to directly interact with Tetratricopeptide Repeat Domain 5 (TTC5), a p53 co-factor, and genetic knockdown of TRIP13 in murine inner medullary collecting duct cells in the presence of hydrogen peroxide showed increased activity of p53 at Serine 15. In all, these studies suggest that insufficient TRIP13 increased the susceptibility of damaged tubular epithelial cells to progress towards apoptotic cell death.


Asunto(s)
ATPasas Asociadas con Actividades Celulares Diversas/deficiencia , Lesión Renal Aguda/patología , Apoptosis , Proteínas de Ciclo Celular/deficiencia , Células Epiteliales/patología , Daño por Reperfusión/patología , Animales , Proteínas de Unión al ADN/metabolismo , Ratones , Unión Proteica , Factores de Transcripción/metabolismo , Proteína p53 Supresora de Tumor/metabolismo
12.
Psychopharmacology (Berl) ; 233(10): 2005-13, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-26900079

RESUMEN

RATIONALE: Gaboxadol is a selective agonist at γ-aminobutyric acidA (GABAA) receptors that contain α4-δ subunits, and it produces anxiolytic and sedative effects. Although adverse effects preclude its clinical use, its mechanism of action suggests that those receptors might provide novel therapeutic targets, particularly for modulators of those GABAA receptor subtypes, by retaining therapeutic effects of gaboxadol and not adverse effects. OBJECTIVES: The current study compared discriminative stimulus effects of gaboxadol with those of modulators acting at GABAA receptors containing α4-δ subunits. MATERIALS: Eight rats discriminated 5.6 mg/kg gaboxadol from vehicle while responding under a fixed - ratio 10 schedule for food. Modulators acting at GABAA receptors containing α4-δ subunits (pregnanolone, ethanol, and flumazenil) and receptors that do not contain those subunits (midazolam) were studied alone; pregnanolone and ethanol were also combined with gaboxadol. In addition, gaboxadol was studied in separate groups discriminating 0.32 mg/kg midazolam, 3.2 mg/kg pregnanolone, or 0.75 g/kg ethanol from vehicle. RESULTS: Gaboxadol produced ≥80 % gaboxadol-lever responding and did not alter rates. No other drug produced, on average, ≥80 % drug-lever responding up to doses that decreased rates, although 1.78 mg/kg midazolam produced 32 % gaboxadol-lever responding. Ethanol and pregnanolone did not enhance the effects of gaboxadol. Rats discriminating midazolam, pregnanolone, or ethanol from vehicle responded predominantly on the vehicle lever after receiving gaboxadol. CONCLUSIONS: Drugs that modulate GABAA receptors containing α4-δ subunits neither mimicked nor enhanced the discriminative stimulus effects of gaboxadol, indicating that at least some effects of gaboxadol are not shared with modulators of that GABAA receptor subtype.


Asunto(s)
Agonistas del GABA/farmacología , Moduladores del GABA/farmacología , Isoxazoles/farmacología , Receptores de GABA-A/metabolismo , Animales , Relación Dosis-Respuesta a Droga , Etanol/farmacología , Flumazenil/farmacología , Masculino , Midazolam/farmacología , Pregnanolona/farmacología , Subunidades de Proteína , Ratas
13.
J Mol Signal ; 10: 6, 2015 Nov 27.
Artículo en Inglés | MEDLINE | ID: mdl-27096004

RESUMEN

Activator of G-protein signaling 3 (AGS3) is an accessory protein that functions to regulate the activation status of heterotrimeric G-protein subunits. To date, however, the downstream signaling pathways regulated by AGS3 remain to be fully elucidated, particularly in renal epithelial cells. In the present study, normal rat kidney (NRK-52E) proximal tubular epithelial cells were genetically modified to regulate the expression of AGS3 to investigate its role on MAPK and mTOR signaling to control epithelial cell number. Knockdown of endogenous AGS3 protein was associated with a reduced phosphorylated form of ERK5 and increased apoptosis as determined by elevated cleaved caspase-3. In the presence of the ERK5 inhibitor, BIX02189, a significant 2-fold change (P < 0.05) in G2/M transition state was detected compared to control conditions. Neither of the other MAPK, ERK1/2 or p38 MAPK, nor another pro-survival pathway, mTOR, was significantly altered by the changes in AGS3 protein levels in the renal epithelial cells. The selective ERK5 inhibitor, BIX02189, was found to dose-dependently reduce NRK cell number by up to 41% (P < 0.05) compared to control cells. In summary, these findings demonstrated that cell viability was regulated by AGS3 and was associated with ERK5 activation in renal epithelial cells.

14.
J Mol Histol ; 46(2): 123-36, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25533045

RESUMEN

Activators of G-protein Signaling (AGS) are a family of accessory proteins that were discovered as modulators of heterotrimeric G-protein subunits. The primary aim of the present study was to localize Group I and II AGS proteins and determine the renal expression profile using immunohistochemistry and quantitative RT-PCR, respectively, during normal and injured states of the kidney. Group I AGS1 was found to be predominantly localized to the proximal tubule, Group II AGS3 and AGS5 were exclusively localized to the distal tubular segments, and Group II AGS6 was ubiquitously expressed in every nephron segment of the rodent kidney. In rat kidneys following ischemia-reperfusion injury (IRI), Group I AGS1 mRNA was dramatically increased after 24 h by fivefold (P < 0.05), whereas Group II AGS3 and AGS4 mRNA was significantly decreased at the same time point (P < 0.05). No significant change in the transcript levels were detected at other time points for any of the AGS genes between control and IRI groups. In polycystic diseased kidneys, mRNA levels for AGS3, AGS4 and AGS6 was significantly increased (P < 0.05) by 75-80 % in PCK rat kidneys. The identification of Group I and II AGS mRNA and protein in the kidney may provide insight into the potential mechanism of action during normal and varying states of renal disease or injury.


Asunto(s)
Proteínas de Unión al GTP/metabolismo , Inhibidores de Disociación de Guanina Nucleótido/metabolismo , Proteínas RGS/metabolismo , Animales , Expresión Génica , Inhibidores de Disociación de Guanina Nucleótido/genética , Riñón/irrigación sanguínea , Riñón/metabolismo , Riñón/patología , Masculino , Ratones , Enfermedades Renales Poliquísticas/metabolismo , Enfermedades Renales Poliquísticas/patología , Transporte de Proteínas , Proteínas RGS/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas Sprague-Dawley , Daño por Reperfusión/metabolismo , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA